WO1996020728A1 - Antitumor agent potentiator comprising il-6 antagonist - Google Patents

Antitumor agent potentiator comprising il-6 antagonist Download PDF

Info

Publication number
WO1996020728A1
WO1996020728A1 PCT/JP1995/002769 JP9502769W WO9620728A1 WO 1996020728 A1 WO1996020728 A1 WO 1996020728A1 JP 9502769 W JP9502769 W JP 9502769W WO 9620728 A1 WO9620728 A1 WO 9620728A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
antagonist
cells
cisplatin
antitumor
Prior art date
Application number
PCT/JP1995/002769
Other languages
French (fr)
Japanese (ja)
Inventor
Osamu Yoshida
Youichi Mizutani
Original Assignee
Chugai Seiyaku Kabushiki Kaisha
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=18336838&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO1996020728(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Chugai Seiyaku Kabushiki Kaisha filed Critical Chugai Seiyaku Kabushiki Kaisha
Priority to US08/860,487 priority Critical patent/US6086874A/en
Priority to ES95942318T priority patent/ES2170815T5/en
Priority to EP95942318A priority patent/EP0800829B2/en
Priority to AT95942318T priority patent/ATE214602T1/en
Priority to KR1019970704446A priority patent/KR100252743B1/en
Priority to CA2209124A priority patent/CA2209124C/en
Priority to DE69525971T priority patent/DE69525971T3/en
Priority to AU43568/96A priority patent/AU704723B2/en
Publication of WO1996020728A1 publication Critical patent/WO1996020728A1/en
Priority to HK98100044A priority patent/HK1001512A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to an antitumor agent potentiator comprising interleukin 6 antagonist which assists and enhances the effect of an antitumor agent in treating a tumor.
  • Renal cell carcinoma is a tumor that is refractory to antitumor agents such as cisplatin, adriamycin and vinblastine (Kakehi, Y. et al., J. Urol., 139, 862- 864, 1988; Kanamaru, et al., J. Natl. Cancer Inst., 81, 844-847, 1989; Teicher, BA et al., Cancer Res., 47, 388-393, 1987).
  • Platinum compounds with antitumor activity, such as cisplatin bind to DNA and inhibit DNA synthesis and cell division (Pinto, AL et al., Biochica et Biophysica Acta, 780, 167-180, 1985).
  • Glutathione-S-transferase-17 ⁇ (GST-r) expression and the effect of cis-bratin due to increased cytoplasmic levels of substances containing sulfidyl groups on the resistance of cell carcinoma to cisplatin. It is thought that suppression, enhancement of DNA repair capacity, or activation of oncogenes such as c-myc are involved in a complex manner (Sklar, MD et al., Cancer Res., 51, 2118-2123, 1991; Mizutani, Y. et al., Cancer in press, 1994; Nakagawa, K. et al., Japan. J. Cancer Res., 79, 301-305, 1988) o
  • Glutathione is tribeptidothiol, an alkylating agent It plays an important role in inactivating DNA-binding substances such as subtilin and repairing cell damage caused by them.
  • One effect of GST-; r is to promote inactivation of the antitumor agent by binding the above antitumor agent to glutathione.
  • IL-16 plays a role in activating the growth of renal cell carcinoma (Miki, S. et al., FEBS Lett., 250, 607-610, 1989; Takenawa,, et al., J. Natl. Cancer Inst., 83, 1668-1672, 1991). ). In addition, it has been reported that serum IL-16 levels are elevated when the prognosis of patients with ⁇ cell carcinoma is poor (Blay, J., et al., Cancer Res., 52, 3317-332, 1992; Tsukamoto, T., et al., J. Urol., 148, 1778-1782, 1992). However, a clear association between IL-16 and the refractory resistance of renal cell carcinoma to antitumor agents has not been established so far, and it is unknown.
  • IL-16 is a multifunctional site-powered protein called B-cell stimulating factor 2 or interferon 2.
  • B-cell stimulating factor 2 or interferon 2.
  • IL-6 was discovered as a differentiation factor involved in the activation of B lymphocyte cells (Hirano, T. et al., Nature 324, 73-76, 1986), and subsequently affected the function of various cells. It has been found to be a multifunctional site kinase (Akira, S. et al., Ad v. In Immunology 54, 1-78, 1993).
  • I_6 transmits its biological activity on cells via two proteins.
  • IL-16R a ligand-binding protein with a molecular weight of about 80 KD to which IL-6 binds.
  • IL-6R exists as a soluble IL-16R (sIL-6R) mainly composed of its extracellular region, in addition to a membrane-bound type expressed through the cell membrane and expressed on the cell membrane.
  • the other is gpl30, which has a molecular weight of approximately 130 KD and is involved in nonligand-bound signal transmission.
  • IL-16 and IL-6R form an IL-6 ZIL-6R complex, followed by another membrane protein, gp By coupling with 130, the biological activity of IL-16 is transmitted to cells (Taga et al., J. Exp. Med., 196, 967, 1987).
  • Platinum compounds such as cis-bratin and antitumor agents such as mitomycin C induce apoptosis in tumor cells, whereas IL-16 suppresses apoptosis induced by antitumour agents (Kerr, J. et al., Cancer 73, 2013-2026, 1994; Sachs, L. et al., Blood 82, 15-21, 1993).
  • antitumor agents such as cisplatin and mitomycin produce cytotoxicity in tumor-producing cells by producing free radicals (Oyanagi, Y. et al., Biochem. Pharmacol. Nakano, H. et al., Biochem. Biophys.
  • IL-6 has a free radical-degrading activity. It is known that the expression of manganese superoxide dismutase (MnSOD) is promoted, and that the 1L-6 antibody suppresses the promoted expression of Mn SOD (Ono, M., et al., Biochem. Biophys. Res. Commun., 182, 1100-1107, 1992; Dougal 1, WC et al., Endocrinology, 129, 2376-2384, 1991) 0
  • An object of the present invention is to provide a new antitumor agent action enhancer that assists and enhances the action of an antitumor agent and increases the sensitivity of tumor cells resistant to treatment with the antitumor agent. More specifically, the present invention provides an antitumor agent action enhancer comprising IL-16 antagonist. More specifically, the present invention provides an action enhancer for a chemotherapeutic agent having an antitumor effect, comprising an IL-16 antagonist. Disclosure of the invention
  • the present inventors have conducted intensive studies on the effects of IL-16 antagonists on changes in the sensitivity of tumor cells to antitumor agents, and have found that IL-6 antagonists such as IL-6 antibody or IL-16R antibody. Increased the sensitivity of tumor cells to the antitumor agent, and a lower dose of the antitumor agent showed a therapeutic effect.
  • the present inventors have found that a therapeutic effect can be exhibited by using an action enhancer containing an antagonist in combination, and have completed the present invention.o
  • the present invention relates to an antitumor agent action enhancer comprising IL-16 antagonist. More specifically, the present invention relates to an agent for enhancing the action of a chemotherapeutic agent having an antitumor effect, which comprises an IL-16 antagonist.
  • an antibody against IL-6 an antibody against IL-6, an antibody against -6R and the like are preferable, and for example, these monoclonal antibodies are preferable.
  • Specific examples of the monoclonal antibody include a PM-1 antibody and a humanized PM-1 antibody.
  • the antitumor agent used in combination with the IL-16 antagonist include chemotherapeutic agents, for example, a platinum compound having antitumor activity, mitomycin C, and the like. Platinum compounds include cisplatin, carboplatin, 254-S, DWA-21 1 4R, NK-121 and the like.
  • Figures 1A and IB show kidney cells in the presence of 0, 0.1, 1 or 10 ⁇ g Zml of cisplatin and IL-6 antibody ( Figure 1A) or IL-6R antibody ( Figure 1B). Shows cytotoxic activity against cancer strain Caki-1. ⁇ indicates only cisplatin, circle indicates coexistence of cisplatin and 0.1 ⁇ g Zml of IL-6 antibody or IL-16R antibody, indicates cisplatin and l ⁇ g noml of IL-6 antibody or IL-6R antibody presence, ginseng represents the cytotoxic activity (%) in the presence of 1 Sea 6 antibody or IL-6R antibody of Shisuburachin and 10/1111.
  • Figures 2A and 2B show the results when mitomycin C and IL-6 antibody ( Figure 2A) or IL-6R antibody ( Figure 2B) coexist at a concentration of 0, 0.1, 1 or 10 g Zml. Shows cytotoxic activity against renal cell carcinoma cell line Caki-1. ⁇ indicates mitomycin C only, garden shows mitomycin C in the presence of 0.1 gZml IL-16 antibody or IL-6R antibody. ⁇ indicates mitomycin C and 1 g / ml IL-16 In the presence of the antibody or the IL-6R antibody, the reference shows the cytotoxic activity () in the presence of the mitomycin C and 10 // g Zml of the IL-6 antibody or the IL-16R antibody.
  • FIGS. 3A and 3B show the results of the renal development in the presence of 0, 0.1, 1 or 10 g / ml of cisplatin and IL-6 antibody (FIG. 3A) or 1L-16R antibody (FIG. 3B). Shows cytotoxic activity against cell carcinoma cell line Caki-1 / DDP.
  • indicates only cisplatin, ⁇ indicates coexistence of cisplatin and 0.1 ⁇ g ml of IL-6 antibody or IL_6R antibody, and ⁇ indicates cisplatin and 1 ⁇ g / ml IL-6 antibody or 1L-16R antibody Indicates the cytotoxic activity (%) in the co-presence of cisplatin and 10 g / ml of the IL-6 antibody or 1L-16R antibody.
  • Figures 4A and 4B show renal cells in the presence of 0, 0.1, 1 or 10 zg Zml of cisplatin and IL-6 antibody ( Figure 4A) or IL-6R antibody ( Figure 4B).
  • Fig. 4 shows cytotoxic activity against cancer cell line ACHN.
  • indicates only cisplatin, ⁇ indicates coexistence of cisplatin and 0.1 ⁇ g Zml of IL-6 antibody or IL-16R antibody, ⁇ indicates cisbratin
  • the pareto shows the cytotoxic activity (%) in the co-presence of cisplatin and 10 gZml of the IL-6 or IL-6R antibody.
  • FIGS. 5A and 5B show the results of the renal development when cisplatin and IL-6 antibody (FIG. 5A) or IL-6R antibody (FIG. 5B) coexist at concentrations of 0, 0.1, 1 or 10 / g Zml.
  • 3 shows cytotoxic activity against cell carcinoma line A704.
  • only cisplatin, fraction: coexistence of cisplatin and 0.1 lz g / ml of IL-6 antibody or IL-6R antibody
  • coexistence of cisplatin and 1 gZml of IL-6 antibody or 1L-16R antibody
  • the lower column shows the cytotoxic activity (%) in the presence of cisplatin and 10 g Zml of IL-16 antibody or 16R antibody.
  • Figures 6A and 6B show patients with vigorous coexistence of cisplatin at 0, 0.1, 1 or 10 g / ml with 1L-6 antibody ( Figure 6A) or 16R antibody ( Figure 6B).
  • 1 shows the cytotoxic activity against fresh renal cell carcinoma obtained from FIG. ⁇ indicates only cisplatin
  • indicates coexistence of 0.1 g Zml of IL-6 antibody or IL-6R antibody
  • indicates coexistence of 1 / g Zml of IL-6 antibody or IL-6R antibody.
  • the lower column shows cytotoxic activity (%) in the presence of cisplatin and 10 / gZml of IL-6 antibody or 1L-6R antibody. .
  • Figures 7A and 7B show the results when 0, 0.1, 1 or 10 ⁇ g Zml of cisplatin and IL-6 antibody (Fig. 7A) or IL-6R antibody (Fig. 7B) coexist vigorously.
  • Cytotoxicity against fresh renal cell carcinoma obtained from patient 2 Show activity.
  • indicates the presence of only cisplatin, and Xin indicates the presence of 0.1 gZm of cisplatin; L-6 antibody or IL-6R antibody.
  • indicates the presence of 1 agZml of IL-16 antibody or IL-6R antibody of cisplatin.
  • Okina shows the cytotoxic activity (%) in the presence of cisplatin and 10 / g ml of IL-6 antibody or IL-6R antibody.
  • Figures 8A and 8B show patients with coexisting 0, 0.1, 1 or 10 ⁇ g Zml of cisplatin and IL-16 antibody ( Figure 8A) or IL-6R antibody ( Figure 8B).
  • 3 shows the cytotoxic activity against fresh renal cell carcinoma obtained from 3.
  • indicates only cisplatin
  • indicates coexistence of cisplatin and 0.1 g of IL-6 antibody or IL-6R antibody
  • indicates that of cisplatin and 1 / zg Zml of IL-16 antibody or IL-16R antibody.
  • Hata shows the cytotoxic activity (%) in the presence of cisplatin and 10 g / ml of IL-16 antibody or IL-6R antibody.
  • FIGS. 9A and 9B show ⁇ cell carcinoma in the presence of lipoplatin at 0, 1, 10 or 100 g Zml at the concentration of —6 antibody (FIG. 9A) or IL-6R antibody (FIG. 9B). Shows cytotoxic activity against strain Caki-1. ⁇ indicates only carbobratin, and Japan indicates the presence of 0.1 lg Zml of I-6 antibody or IL-6R antibody in the country. ⁇ indicates 1 ug / ml of 6 antibody or IL-6R antibody with carboplatin. In the coexistence, Hata shows the cytotoxic activity (%) in the coexistence of calpoplatin and 10 8 11) 1 1-6 antibody or IL-16R antibody.
  • FIG. 10 indicates GST-in renal cell carcinoma cell line Caki_1 treated with culture medium (control), 10iz g Zml of cis-bratin, 10 / g Zml of IL-16 antibody or IL-16R antibody.
  • FIG. 7 shows the results of Northern blotting of total Caki-1 RNA using a GST- ⁇ cDNA probe for examining 7 ⁇ mRNA expression. Lane 1 was treated with culture medium only, lane 2 was treated with cisplatin, lane 3 was treated with IL-6 antibody, and lane 4 was treated with 1-6R antibody .
  • B in FIG. 10 is a diagram of a gel used for Northern plots, which was stained with ethidium umide. Each lane indicates the presence of RNA. Specific description
  • the antitumor agent-enhancing agent comprising 1L-6 antagonist of the present invention enhances the antitumor effect when used in combination with an antitumor agent in treating a tumor. In addition, it has the effect of reducing the required dose of an antitumor agent and increasing the sensitivity of the antitumor agent even to a treatment-resistant tumor for which a therapeutic effect is not recognized by ordinary chemotherapy.
  • the anti-tumor agent whose antitumor effect is enhanced by the action enhancer of the present invention is a chemotherapeutic agent that acts on tumor cells to suppress the growth and proliferation of tumor cells and has a therapeutic effect on tumors.
  • Chemotherapeutic agents include alkylating agents, metabolic antagonists, antitumor antibiotics, plant-derived alkyloids, formone therapeutic agents, and platinum compounds.
  • a platinum compound having an antitumor effect of mitomycin C ⁇ which is an antitumor agent, is particularly preferred.
  • Platinum compounds have a platinum atom and form a complex with other atoms.
  • platinum compounds having antitumor activity include cisplatin (cis-diamminedich1 orop1 atinum (11), having the following structural formula).
  • NK-121 ((R) -cis-2-methy 1 -1,4-butane.diamine (1, 1-cyclobutaned icarboxylato) plat inum (II), having the following structural formula), Okisariburachin (oxaliplatin; USAN, Oxalato (trans -1, 2-cyc lohexanediamine) platinum (11), have a structural formula below _ 9),
  • TRK-710 ((alpha-acetyl-gamma-methyl tetronate) z- (I-1, 2-diaminocyclohexane) platinum (II), having the following structural formula)
  • Etc. are known.
  • platinum compounds cisplatin, carboplatin and DWA-2114R are particularly preferred.
  • antitumor agents are formulated in a usual manner.
  • a platinum compound if necessary, it is mixed with an adjuvant and a simple substance used as a drug, and used orally or parenterally, preferably as an injection.
  • an injection it is advisable to mix it with distilled water or a salt solution such as sodium chloride or potassium chloride, a glucose solution or a physiological saline solution.
  • the amount of the antitumor agent in these preparations is desired to be a convenient unit dose depending on the age and condition of the patient. For example, when used for the treatment of tumors in adults, 10 to 2000 mgZm once a day 2 (volume area) may be administered, and depending on the dose, continuous administration for 5 days or a drug holiday of 114 weeks between administrations may be provided.
  • Tumor cells having an antitumor effect by the action enhancer of the present invention are tumors having IL-6R and exhibiting proliferation and Z or treatment resistance using IL-6 as one physiologically active substance.
  • Such tumors include renal cell carcinoma (Miki, S. et al., FEBS Letter, 250, 607-610, 1989) and myeloma (Kawano, M. et al., Nature, 332, 83-85, 1988). ), Ovarian cancer (Kobayash i, H. et al., Proceedings of the 53rd Annual Meeting of the Japanese Cancer Society, 271 pages, 874, 1994), EB virus-infected B lymphoma (Tosata, G. et al., J. Virol.
  • the IL_6 antagonist used in the present invention may be of any origin as long as it blocks signal transmission by IL-6 and inhibits the biological activity of 1L-16.
  • IL-6 antagonist IL-6 Body, IL-6R antibody, gpl30 antibody, IL-6 variant, antisense oligonucleotide of IL-6R, partial peptide of IL-16 or IL-6R, and the like.
  • Antibodies used as 1L-16 antagonists in the present invention may be of any origin and type (monoclonal, polyclonal). In particular, a monoclonal antibody derived from a mammal is preferred.
  • IL-6 antibody, IL-6R or gpl30 these antibodies inhibit the binding of IL-6 to IL-6R or IL-6R and gpl30, block 1L_6 signaling, and Is an antibody that inhibits the biological activity of
  • the animal species of the cell producing the monoclonal antibody is not particularly limited as long as it is a mammal, and may be derived from a human antibody or a mammal other than human.
  • a monoclonal antibody derived from a mammal other than human a monoclonal antibody derived from a rabbit or a rodent is preferred because of its ease of preparation.
  • the rodent is not particularly limited, but mouse, rat, hamster and the like are preferably exemplified.
  • Such antibodies include MH166 antibody (Matsuda et al., Eur. J. Immunol. 18: 951-956, 1988) and SK2 antibody (Sato et al., 21st Annual Meeting of the Immunological Society of Japan). Scientific Records, 21: 116, 1991).
  • Examples of the IL-6R antibody include PM-1 antibody (Hirata et al., J. Immunol. 143: 2900-2906, 1989), AUK12-20 antibody, AUK64-7 antibody, and AUK146-5-15 antibody (International Patent Application Publication).
  • the IL-6R antibody is particularly preferable, and specific examples thereof include the above-mentioned PM-1 antibody.
  • a monoclonal antibody can be basically produced using a known technique as follows. That is, IL-6, IL-6R or gpl30 It is used as a sensitizing antigen, and is immunized according to a conventional immunization method, and the obtained immune cells are fused with a known parent cell by a normal cell fusion method, and are then subjected to a normal screening method. It can be prepared by screening monoclonal antibody-producing cells.
  • the sensitizing antigen is preferably a human-derived antigen.
  • the human sensitizing antigen is a human IL-16 which is disclosed in Hirano et al., Nature, 324: 73, 1986. It is obtained by using gene sequences. After the human 6 gene sequence is inserted into a known expression vector system to transform an appropriate host cell, the desired IL-6 protein is isolated from the host cell or the culture supernatant. After purification, the purified IL-6 protein may be used as a sensitizing antigen.
  • an IL-6R protein can be obtained by using the gene sequence disclosed in European Patent Application Publication No. EP325474 and following the same method as for human IL-16.
  • IL-6R There are two types of IL-6R: those expressed on the cell membrane and those soluble off the cell membrane (sIL-6R) (Yasukawa et al., J. Biochem., 108, 673-676, 1990).
  • sIL-6R is composed mainly of the extracellular region of IL-16R bound to the cell membrane, and differs from membrane-bound IL-16R in that the transmembrane region or the transmembrane region and the intracellular region are defective. Is different.
  • gpl30 protein can be obtained by using the gene sequence disclosed in European Patent Application Publication No. EP411946 and following the same method as for IL-6.
  • the mammal to be immunized with the sensitizing antigen is not particularly limited, but is preferably selected in consideration of compatibility with the parent cell used for cell fusion. For this, mice, rats, hamsters, egrets, etc. are used. Immunization of an animal with a sensitizing antigen is performed according to a known method. For example, as a general method, a sensitizing antigen is injected into a mammal intraperitoneally or subcutaneously. Specifically, sensitizing antigen
  • PBS Phosphate-Buffered Saline
  • physiological saline physiological saline
  • an appropriate carrier can be used at the time of immunization with the sensitizing antigen.
  • immune cells are removed from the mammal and subjected to cell fusion. Preferred immune cells are particularly preferred. Splenocytes. Mammalian myeloma cells as the other parent cells fused with the immune cells are already known in various cell lines, for example, P3 (P3x63Ag8.653) (J. Immunol. 123: 1548, 1978), p3-Ul (Current Topics in Micro-biology and Immunology 81: 1-7, 1978), NS-1 (Eur. J. Immunol.
  • Cell fusion between the immune cells and myeoma cells is basically performed according to a known method, for example, the method of Milstein et al. (Milstein et al., Methods Enzymol. 73: 3-46, 1981). It can be carried out.
  • the cell fusion is performed, for example, in a normal nutrient culture in the presence of a cell fusion promoter.
  • a cell fusion promoter for example, polyethylene glycol (PEG), Sendai virus (HVJ) or the like is used, and if necessary, an auxiliary agent such as dimethyl sulfoxide is added to enhance the fusion efficiency.
  • PEG polyethylene glycol
  • HVJ Sendai virus
  • the ratio of the use of the immune cells to the myeloma cells is preferably, for example, 11 to 10 times the number of the immune cells to the myeoma cells.
  • the culture medium used for cell fusion include RPM11640 culture medium and MEM culture medium suitable for the proliferation of the myeloma cell line, and other ordinary cultures used for cell culture of this type.
  • a fluid can be used, and a serum replacement fluid such as fetal calf serum (FCS) can also be used in combination.
  • FCS fetal calf serum
  • a predetermined amount of the immune cells and myeloma cells is mixed well in the culture medium, and a PEG solution previously heated to about 37 ° C., for example, an average molecular weight of about 1,000 to 6,000
  • the desired fused cells are formed by adding and mixing the PEG solution of the above at a concentration of 30-60% (wZv). Subsequently, by repeating the operation of successively adding an appropriate culture solution and centrifuging to remove the supernatant, a cell fusion agent or the like unfavorable for the growth of the hybridoma can be removed.
  • the hybridoma is selected by culturing it in a normal selective culture solution, for example, a HAT culture solution (a culture solution containing hypoxanthine, aminobuterin and thymidine). Culture in the HAT culture medium is continued for a period of time sufficient to kill cells other than the target hybridoma (non-fused cells), usually several days to several weeks. Next, a conventional limiting dilution method is performed, and screening and monocloning of the hybridoma producing the desired antibody are performed.
  • a normal selective culture solution for example, a HAT culture solution (a culture solution containing hypoxanthine, aminobuterin and thymidine). Culture in the HAT culture medium is continued for a period of time sufficient to kill cells other than the target hybridoma (non-fused cells), usually several days to several weeks.
  • a conventional limiting dilution method is performed, and screening and monocloning of the hybridoma producing the desired antibody are performed.
  • the hybridoma producing the monoclonal antibody thus produced can be subcultured in a normal culture medium, and can be stored for a long time in liquid nitrogen. .
  • a method of culturing the hybridoma according to an ordinary method and obtaining the culture supernatant, or transferring the hybridoma to a mammal compatible therewith Injection, proliferation and ascites are used.
  • the former method is suitable for obtaining high-purity antibodies, while the latter method is suitable for mass production of antibodies.
  • Monoclonal antibodies include not only those obtained from hybridomas produced by cell fusion of antibody-producing cells obtained by immunization with an antigen, but also those obtained by cloning an antibody gene and incorporating it into an appropriate vector. Can be used as a monoclonal antibody produced by using a gene recombination technique by introducing the gene into a known cell line, for example, COS, CH0, etc. (for example, Vandamme, A-M. Et al., Eur J. Biochem., 192, 767-775, 1990).
  • the monoclonal antibody obtained by the above-mentioned method can be purified to a high degree of purity using ordinary purification means such as a salting-out method, a gel filtration method, and an affinity chromatography method. It can be purified.
  • the monoclonal antibodies thus prepared can be obtained by ordinary immunological means such as radioimmunoassay (RIA), enzyme immunoassay (E1A, ELISA), and immunofluorescence assay (Immunofluorescence analysis). It can be confirmed that the antigen is recognized with high sensitivity and high accuracy.
  • immunological means such as radioimmunoassay (RIA), enzyme immunoassay (E1A, ELISA), and immunofluorescence assay (Immunofluorescence analysis). It can be confirmed that the antigen is recognized with high sensitivity and high accuracy.
  • the monoclonal antibody used in the present invention is not limited to the monoclonal antibody produced by the hybridoma, but may be artificially modified for the purpose of, for example, reducing the antigenicity to humans. What you do is better.
  • a chimeric antibody comprising a variable region of a mammalian non-human, eg, mouse, monoclonal antibody and a constant region of a human antibody can be used. It can be produced using known methods for producing chimeric antibodies, particularly using gene recombination techniques.
  • a reshaped human antibody can be used in the present invention. This is obtained by transplanting the complementarity-determining regions of a mammal other than a human, for example, a mouse antibody, into the complementarity-determining regions of a human antibody. Is also known. Using the known method, a reshaped human antibody useful in the present invention can be obtained, and a preferred example thereof is a reshaped PM-1 antibody (for example, an international patent) (See Application Publication No. W092-19759).
  • amino acid in the framework (FR) region of the variable region of the antibody may be replaced so that the complementarity determining region of the reshaped human antibody forms an appropriate antigen binding site.
  • antibody fragments such as F (ab ') 2 , Fab or Fv, and Fv of H and L chains, with appropriate linkers, as long as they bind to antigen and inhibit the activity of IL-6
  • scFv single-glutinin Fv
  • scFv is formed by linking the H chain V region and L chain V region of an antibody.
  • the H chain V region and the L chain V region are linked via a linker, preferably a peptide linker (Huston, JS et al., Proc. Natl. Acad. Sci. USA, 85, 5879-5883, 1988).
  • the H chain V region and L chain V region in scFv may be derived from any of the antibodies described above. These V regions are preferably connected by peptide linkers.
  • the peptide linker for example, any single-chain peptide consisting of amino acid residues 12 to 19 is used.
  • the scFv-encoding DNA is a DNA encoding the H chain or the H chain V region of the antibody, and a DNA encoding the L chain or the L chain V region.
  • the DNA portion encoding the desired amino acid sequence is amplified by the PCR method using a pair of primers defining both ends, and then a portion of the peptide linker is also encoded. It is obtained by combining and widening a pair of primers that define DNA and its both ends so as to be linked to an H chain and an L chain, respectively.
  • DNAs encoding scFv are prepared, an expression vector containing them and a host transformed by the expression vector can be obtained according to a conventional method.
  • the scFv can be obtained according to a conventional method.
  • the scFv is superior to the tissue transferability of the antibody molecule, and is expected to have the same function as the reshaped human antibody.
  • the IL-16 variants used in the present invention include those disclosed in Brakenhoff et al., J. Biol. Chem. 269: 86-93, 1994 or Savino et al., EMBO J. 13: 1357-1367, 1994. Is mentioned.
  • the IL-6R can maintain its IL-6R binding activity while maintaining its IL-6R binding activity.
  • a mutation such as substitution, deletion, or human mutation
  • 1L-6 as its origin is not limited to animal species as long as it has the above properties, but it is preferable to use human origin in consideration of antigenicity.
  • the amino acid sequence of IL-16 can be obtained by using a known molecular modeling program such as WHATIF (Vriend et al., J. Mol. Graphics, 8: 52-56, 1990). This is done by predicting the secondary structure and assessing the overall effect of the mutated amino acid residue.
  • the vector containing the nucleotide sequence encoding the human IL-16 gene is used as a template for PCR (polymerase chain reaction).
  • PCR polymerase chain reaction
  • a gene encoding a 1L-6 variant can be obtained.
  • This is inserted into an appropriate expression vector if necessary, expressed in E. coli cells or mammalian cells, and isolated or purified as it is contained in the culture supernatant or by a conventional method. Binding activity to IL-6 and IL-6 The neutralizing activity of the signal transmission can be evaluated.
  • the IL-16 partial peptide or IL-6R partial peptide used in the present invention binds to IL-6R or IL-6, respectively, as long as it has no IL-16 activity transfer action. Regardless of the sequence of the fragment. See US Patent Publication US5210 075 and European Patent Publication No. 6-1126 for IL-6 and IL-6R partial peptides. For the IL-6R antisense oligonucleotide, see Japanese Patent Application No. 5-300338.
  • the antitumor agent potentiator comprising the IL-6 antagonist of the present invention has an IL-16R as long as it blocks 1L-6 signal transduction and captures and enhances the action of the antitumor agent.
  • IL-6 can be effectively used as a physiologically active substance for the treatment of any tumor that shows growth and no resistance or treatment resistance.
  • the potentiator of the antitumor agent comprising the IL-16 antagonist of the present invention is preferably administered parenterally, for example, by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, etc. It can be administered steadily. Furthermore, it can be in the form of a pharmaceutical composition kit with at least one pharmaceutical carrier or diluent.
  • the dose of the antitumor agent potentiator comprising IL-16 antagonist of the present invention to a human varies depending on the patient's condition, age and administration method, but it is necessary to select an appropriate amount as appropriate.
  • an appropriate amount for example, in the case of IL-6R antibody, no more than four divided doses can be selected in a range of approximately 11 mg / l mg of patients. It can also be administered at a dose of 11 mg / kgZweek.
  • the dose of the antitumor agent potentiator comprising the IL-6 antagonist of the present invention is not limited to these doses.
  • Antitumor agent action enhancer comprising 16 antagonist of the present invention
  • injectable preparations are prepared by dissolving purified IL-16 gonist in a solvent such as physiological saline, buffer, glucose solution, etc., and adding an anti-adsorption agent such as Tween80, gelatin Or serum albumin (HSA) or lyophilized for reconstitution before use.
  • an excipient for freeze-drying for example, sugar alcohols and sugars such as mannitol and glucose can be used.
  • Human renal cell carcinoma cell lines Caki-1 and Caki-1 are sublines of cis-bratin-resistant cell line Caki-1 ZDDP, human cell carcinoma ACHN, and human renal cell carcinoma A704 (Giard, DJ et al., J. Nat. 1. Cancer Inst. 51, 1417-1423, 1973) was added to 25 mM HEPES, 2 mM L-octylamine, 1% non-essent ial amino acid, 100 U Zml penicillin, 100 g / ml Streptmycin and 10% heat-inactivated. In a RPMI 1640 culture medium (above, manufactured by Gibco) containing fetal serum (FBS) (hereafter referred to as complete culture medium), on a plastic dish. The cells were cultured to form a monolayer.
  • FBS fetal serum
  • neoplastic tumor cells were obtained from renal cell carcinoma patients.
  • renal cell carcinoma tumor tissue was obtained.
  • SmgZml collagenase manufactured by Sigma Chemical Co.
  • the cell suspension was placed on a discontinuous gradient consisting of 2 ml each of 100%, 80% and 50% fico-hypaque in a 15 ml plastic tube.
  • the layers were overlaid and centrifuged at 400 ⁇ g for 30 minutes. Remove the lymphocyte-enriched monocyte layer from the 100% layer and obtain tumor cells and mesothelial cells from 80% eyebrows.To prevent contamination with other cells, complete culture in a 15 ml plastic tube.
  • the cell suspension, enriched in tumor cells was overlaid on a discontinuous gradient consisting of 3 ml each of 25%, 15%, 10% Percoll and centrifuged at room temperature at 25 ⁇ g for 7 minutes. Tumor cells separated from lymphocytes were obtained from the bottom of the tube. After the obtained tumor cells were washed and suspended in a complete culture solution, the survival of the tumor cells was confirmed by trypan blue staining. These tumor cells thus prepared were used in the following experiments.
  • IL IL-6 is present in the culture supernatant of cell tumor cell lines Caki-1 and Caki-1 / DDP, ACHN, A704 and 704 cell carcinoma patients (No. 1 to 3). Was assayed by the BLISA (enzyme-linked immunosorbent assay; method).
  • a 96-well EUSA plate was supplemented with 100-1 IL-16 antibody, and the ELISA plate was coated with the IL-6 antibody at least. These plates were stored at 4 ° C for up to 4 weeks before use. Plates coated with IL-16 antibody were washed three times and blocked for 1 hour with ELISA PBS containing 1% BSA (perforated serum albumin). After two washes, Escherichia coli-derived recombinant IL-16 (Yasukawa et al., Biotechnol. Lett., 12, 419, 1990) was added to each pellet as a 100 ⁇ 1 tumor cell culture supernatant or control.
  • RCC cells IL-1-6 degree (pgZml; average soil standard deviation)
  • Fresh RCC cells (Patient No. 1) 1236 ⁇ 71
  • Fresh RCC cells (Patient No. 2) 42 ⁇ 4
  • Fresh RCC cells (Patient No. 3) 2579 ⁇ 219
  • cisplatin cis-diamminedi chloroplatinum (II)
  • mitomycin C MMC
  • ADR adriamycin
  • VBL vinblastine
  • 5 — phnoreolouracinole 5 — IL-6 antibody or 1L on the sensitivity of each renal cell Caki-1, Caki-1 / DDP, ACHN, A704 and fresh tumor cells from the patient (No. 1-3) to F. luorouraci 1;
  • MTT method Mizutani, Y. et al., Cancer 73, 730-737, 1994
  • IL-6 antibody Matsuda, T. et al., Eur. J. Immunol., 18, 951-956, 1988
  • IL-6R antibody Hirata, Y. et al., J.
  • Example 3 IL-16 antibody or IL-1 related to intracellular accumulation of antitumor agent
  • the medium is then removed and the cells are washed three times with RPMI 1640 medium did.
  • the intracellular accumulation of cisplatin was determined by the frameless atomic absorption spectrometry method (Daley-Tates, PT et al., Biochem. Pharmacol., 34, 2263-2369; Riley, CM et al., Analytical Biochem., 124, 167). -179, 1982).
  • Zeeman Z-8000 Zeeman Z-8000 (Zeeman z-8000 Spectrophotometer, manufactured by Hitachi Co. Ltd.) was used.
  • the intracellular accumulation of 5-FU was measured by gas chromatography-mass spectrometry (Marunaka, T., et al., J. Pharm.
  • Control Control (Culture medium) Control Ab Anti-IL-6mAb Anti-IL-6R mAb CDDP 0.28 ⁇ 0.05 0.27 ⁇ 0.02 0.26 ⁇ 0.05 0.27 ⁇ 0.02 5—FU 1.48 ⁇ 0.32 1.57 ⁇ 0.45 1.50 ⁇ 0.19 1.63 ⁇ 0.31 Numerical values of three experiments Calculated from average data (average soil standard deviation)
  • RNAZ lanes Effect of cisplatin, IL-6 antibody or 16R antibody on Caki-1 cells as control medium, 10 g Zml cisplatin, 10 ⁇ g / ml IL-6 antibody or IL-1R
  • the cells were cultured with the 6R antibody for 4 hours.
  • the total RNA of the cells was prepared by the method of Mizutani, Y, et al. (Cancer 73, 730-737, 1994), and 200 mM mops (M0PS; 3-[N-morpholy) was used to obtain 10 g RNAZ lanes.
  • RNA was transferred to Biodyne A membrane (Poll) in a 20X SSC solution containing 3 M NaCl and 0.3 M sodium citrate (pH 7.0). 50—100 ng of GST—7 ⁇ cDNA probe (Nakagawa, K. et al., J. Biol. Cem. 265, 4296-4301, 1990) labeled with 32 P—dCTP (NEN) using the random oligo primer extension method did.
  • the nylon membrane onto which the RNA was transferred was cross-linked with ultraviolet light and hybridized with the probe. The results are shown in FIG.
  • Cisplatin did not affect GST-7 ⁇ mRNA expression in Caki-1 cells.
  • the addition of IL-6 antibody or IL-6R antibody reduced the expression of GST-7 mRNA.
  • an IL-16 antagonist such as an IL-6 antibody or an IL-6R antibody
  • the sensitivity of the tumor cell to the antitumor agent was observed at a lower dose, and the IL-16 antagonist was inhibited by the antitumor agent. It is confirmed that the combination results are exhibited by the tumor agent. Furthermore, it is demonstrated that tumor cells that are resistant to treatment with an antitumor agent can be treated as a result of enhancing the sensitivity of the antitumor agent to IL-16 antagonist.
  • the potentiator of the antitumor agent comprising IL-6 antagonist of the present invention can reduce the toxicity of the antitumor agent to tissues by reducing the required dose of the antitumor agent. Therefore, it is expected as an action enhancer of an antitumor agent.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Inorganic Chemistry (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Mycology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A potentiator for antitumor agents, for example, platinum compounds such as cisplatin and carboplatin, mitomycin C, etc., which comprises an interleukin-6 (IL-6) antagonist such as an antibody against IL-6, an antibody against IL-6 receptor, an antibody against protein gp130, etc.

Description

明 細 誊  明
IL一 6 アンタゴニス トを含んでなる抗腫瘍剤の作用増強剤 技術分野 Field of action of antitumor agents comprising IL-16 antagonist
本発明は、 腫瘍の治療において抗腫瘍剤の作用を補助、 増強する イ ンターロイキン 6 アンタゴニス トを含んでなる抗腫瘍剤の作用增 強剤に関する。  The present invention relates to an antitumor agent potentiator comprising interleukin 6 antagonist which assists and enhances the effect of an antitumor agent in treating a tumor.
背景技術 Background art
ヒ 卜腫瘍の化学療法には、 これまでアルキル化剤、 代謝拮抗剤、 抗腫瘍抗生剤、 白金化合物等が用いられてきた。 これらの抗腫瘍剤 を単独で用いても顕著な治療効果が認められない場合、 複数の抗腫 瘍剤を併用する治療法が考えられてきた (Frei, E. Ill, Cancer R es., 32, 2593-2607, 1992) 。 腫瘍細胞は、 抗腫瘍剤に様々な感受 性を有し、 あるものは抗腫瘍剤に対して治療抵抗性を示すことが知 られている (Magrath, I. , New Directions in Cancer Treatment, 1989, Springer-Verlag) 。 腫瘍細胞の治療抵抗性獲得には、 マル チ ドラ ッ グレジスタ ンス (multi- drug resistance ; MDR)の発現 ( Tsuruo, T. ら、 Cancer Res. , 42, 4730-4733, 1982) 、 抗腫瘍剤の 細胞内取込みの減少 (Sherman, S. E. ら、 Science, 230, 412, 198 5)、 DNA 修復活性の增加 (Borc , R. F. , Metabolism and Action o f Anticancer Drugs. Powis, G. & Prough, R.編、 Taylor & Franc is, London, 1987, 163- 193)または細胞内での抗腫瘍剤の不活化の 促進 (Teicher, B. A. ら、 Cancer Res., 46, 4379, 1986)等がその 原因であるといわれている。 このような場合、 抗腫瘍剤を投与する だけでは予期した治療効果が認められない場合が多い。 腎細胞癌 (renal cell carcinoma) は、 シスブラチン、 ア ド リ ア マイ シンおよびビンブラスチンなどの抗腫瘍剤に治療抵抗性を示す 腫瘍である (Kakehi, Y. ら、 J. Urol., 139, 862-864, 1988 ; Kana maru, ら、 J. Natl. Cancer Inst. , 81, 844-847, 1989 ; Teicher , B.A. ら、 Cancer Res. , 47, 388-393, 1987) 。 シスブラチンなど の抗腫瘍作用を有する白金化合物は、 DNA に結合し、 DNA の合成お よび細胞の分裂を阻害する (Pinto, A. L. ら、 Biochica et Biophy sica Acta, 780, 167-180, 1985)。 For chemotherapy of human tumors, alkylating agents, antimetabolites, antitumor antibiotics, platinum compounds and the like have been used. If no significant therapeutic effect is observed even when these antitumor agents are used alone, a treatment method combining multiple antitumor agents has been considered (Frei, E. Ill, Cancer Res., 32). , 2593-2607, 1992). Tumor cells have various sensitivities to antineoplastic drugs, and some are known to exhibit resistance to antitumor drugs (Magrath, I., New Directions in Cancer Treatment, 1989, Springer-Verlag). Expression of multi-drug resistance (MDR) (Tsuruo, T. et al., Cancer Res., 42, 4730-4733, 1982) Decreased cellular uptake (Sherman, SE et al., Science, 230, 412, 1985), increased DNA repair activity (Borc, RF, Metabolism and Action of Anticancer Drugs. Powis, G. & Prough, R., Taylor & Francis, London, 1987, 163-193) or promotion of inactivation of antitumor agents in cells (Teicher, BA et al., Cancer Res., 46, 4379, 1986). I have. In such cases, administration of an antitumor agent alone often does not provide the expected therapeutic effect. Renal cell carcinoma is a tumor that is refractory to antitumor agents such as cisplatin, adriamycin and vinblastine (Kakehi, Y. et al., J. Urol., 139, 862- 864, 1988; Kanamaru, et al., J. Natl. Cancer Inst., 81, 844-847, 1989; Teicher, BA et al., Cancer Res., 47, 388-393, 1987). Platinum compounds with antitumor activity, such as cisplatin, bind to DNA and inhibit DNA synthesis and cell division (Pinto, AL et al., Biochica et Biophysica Acta, 780, 167-180, 1985).
肾細胞癌のシスブラチンに対する治療抵抗性には、 グルタチオン - S ト ラ ンスフ エ レース一 7Γ (GST— r ) の発現、 スルフ ィ ド リ ル 基を含有する物質の細胞質内レベル上昇による シスブラチ ンの作用 抑制、 DNA 修復能上昇あるいは c —myc などの発ガン遗伝子の活性 化等が複雑に関与していると考えられている (Sklar, M. D. ら、 Ca ncer Res., 51, 2118 - 2123, 1991 ; Mi zutani, Y. ら、 Cancer in press, 1994 ; Nakagawa, K. ら、 Japan. J. Cancer Res. , 79, 301-3 05, 1988) o  肾 Glutathione-S-transferase-17Γ (GST-r) expression and the effect of cis-bratin due to increased cytoplasmic levels of substances containing sulfidyl groups on the resistance of cell carcinoma to cisplatin. It is thought that suppression, enhancement of DNA repair capacity, or activation of oncogenes such as c-myc are involved in a complex manner (Sklar, MD et al., Cancer Res., 51, 2118-2123, 1991; Mizutani, Y. et al., Cancer in press, 1994; Nakagawa, K. et al., Japan. J. Cancer Res., 79, 301-305, 1988) o
また、 腫瘍細胞での膜透過輸送能の変化は、 細胞内のシスプラチ ンの取込み減少をもたら し、 シスブラチンに対する治療抵抗性を增 すといわれている (Richon, V. ら、 Cancer Res. , 47, 2056-2061, 1987 ; Waud, W. R. ら、 Cancer Res. , 47, 6549-6555, 1987) 。 ス ルフ ィ ドリル基を含む物質と して、 哺乳類動物細胞内に最も豊富に 存在するダルタチオンは、 細胞内でシスブラチンを不活化するこ と が報告されており、 ある種の腫瘍において細胞内のグルタチオンお よびメ タロチォネィ ンレベルが高く なっているこ とが示された (Hr omas, R. A. ら、 Cancer LETT. , 34, 9 - 13, 1987 ; Taylor, D. M. ら 、 Eur. J. Cancer, 12, 249-254, 1976)。  In addition, it is said that changes in the transmembrane transport ability of tumor cells result in decreased uptake of intracellular cisplatin and increase resistance to treatment with cisplatin (Richon, V. et al., Cancer Res., 47, 2056-2061, 1987; Waud, WR et al., Cancer Res., 47, 6549-6555, 1987). Daltathione, which is the most abundant substance in mammalian cells as a substance containing a sulfidyl group, has been reported to inactivate cisbratin in cells, and intracellular glutathione in some tumors. And increased levels of metallothionein (Hromas, RA et al., Cancer LETT., 34, 9-13, 1987; Taylor, DM et al., Eur. J. Cancer, 12, 249- 254, 1976).
グルタチオンは ト リベプチ ドチオールであり、 アルキル化剤ゃシ スブラチンのような DNA 結合物質の不活化およびこれらによる細胞 障害の修復に重要な役割を担っている。 GST— ; rの有する一つの作 用は、 上記のような抗腫瘍剤をグルタチオンに結合させることによ り抗腫瘍剤の不活化を促進することである。 Glutathione is tribeptidothiol, an alkylating agent It plays an important role in inactivating DNA-binding substances such as subtilin and repairing cell damage caused by them. One effect of GST-; r is to promote inactivation of the antitumor agent by binding the above antitumor agent to glutathione.
肾細胞癌は、 イ ンターロイキン 6 ( IL- 6 ) を産生し、 IL一 6 レ セプター (IL一 6R) を発現していることから、 腎細胞癌の増殖活性 化に IL一 6が何らかの役割を担っていることが示唆されている (Mi ki, S. ら、 FEBS Lett. , 250, 607 - 610, 1989 ; Takenawa, , ら、 J. Natl. Cancer Inst. , 83, 1668-1672, 1991) 。 さらに、 肾細胞癌 患者の治療予後が悪い場合、 血清中の IL一 6 レベルが上昇している ことが報告されている (Blay, J. , ら、 Cancer Res., 52, 3317-33 22, 1992 ; Tsukamoto, T., ら、 J. Urol. , 148, 1778-1782, 1992) 。 しかしながら、 IL一 6 と腎細胞癌の抗腫瘍剤に対する治療抵抗性 とはこれまでに明確な関連づけがなされておらず不明であった。  肾 Since cell carcinoma produces interleukin 6 (IL-6) and expresses IL-16 receptor (IL-16R), IL-16 plays a role in activating the growth of renal cell carcinoma (Miki, S. et al., FEBS Lett., 250, 607-610, 1989; Takenawa,, et al., J. Natl. Cancer Inst., 83, 1668-1672, 1991). ). In addition, it has been reported that serum IL-16 levels are elevated when the prognosis of patients with 肾 cell carcinoma is poor (Blay, J., et al., Cancer Res., 52, 3317-332, 1992; Tsukamoto, T., et al., J. Urol., 148, 1778-1782, 1992). However, a clear association between IL-16 and the refractory resistance of renal cell carcinoma to antitumor agents has not been established so far, and it is unknown.
IL一 6 は B細胞刺激因子 2あるいはインタ一フ ュ ロ ン 2 と呼称 された、 多機能サイ ト力イ ンである。 IL— 6 は B リ ンパ球系細胞の 活性化に関与する分化因子と して発見され (Hirano, T. ら、 Nature 324, 73-76, 1986)、 その後、 種々の細胞の機能に影響を及ぼす多 機能サイ トカイ ンであることが明らかとなつた (Akira, S. ら、 Ad v. in Immunology 54, 1-78, 1993) 。 Iし _ 6 は、 細胞上で二種のタ ンパク質を介してその生物学的活性を伝達する。  IL-16 is a multifunctional site-powered protein called B-cell stimulating factor 2 or interferon 2. IL-6 was discovered as a differentiation factor involved in the activation of B lymphocyte cells (Hirano, T. et al., Nature 324, 73-76, 1986), and subsequently affected the function of various cells. It has been found to be a multifunctional site kinase (Akira, S. et al., Ad v. In Immunology 54, 1-78, 1993). I_6 transmits its biological activity on cells via two proteins.
一つは、 IL— 6が結合する分子量約 80KDのリガン ド結合性タンパ ク質、 IL一 6Rである。 IL— 6Rは、 細胞膜を貫通して細胞膜上に発現 する膜結合型の他に、 主にその細胞外領域からなる可溶性 IL一 6R(s IL-6R) と しても存在する。 もう一つは非リガン ド結合性のシグナ ル伝達に係わる分子量約 130KD の gpl30 である。 IL一 6 と IL-6Rは IL- 6 ZIL— 6R複合体を形成し、 次いでもう一つの膜タンパク質 gp 130 と桔合することにより、 IL一 6 の生物学的活性が細胞に伝達さ れる (Tagaら、 J. Exp. Med., 196, 967, 1987) 。 One is IL-16R, a ligand-binding protein with a molecular weight of about 80 KD to which IL-6 binds. IL-6R exists as a soluble IL-16R (sIL-6R) mainly composed of its extracellular region, in addition to a membrane-bound type expressed through the cell membrane and expressed on the cell membrane. The other is gpl30, which has a molecular weight of approximately 130 KD and is involved in nonligand-bound signal transmission. IL-16 and IL-6R form an IL-6 ZIL-6R complex, followed by another membrane protein, gp By coupling with 130, the biological activity of IL-16 is transmitted to cells (Taga et al., J. Exp. Med., 196, 967, 1987).
シスブラチ ンのような白金化合物や、 マイ トマイ シ ン C といった 抗腫瘍剤は、 腫瘍細胞にアポ ト一シスを誘導するが、 IL一 6 は抗腫 癘剤により誘導されたアポ トーシスを抑制することが報告されてい る (Kerr, J. ら、 Cancer 73, 2013-2026, 1994 ; Sachs, L. ら、 Bl ood 82, 15-21, 1993)。 また、 シスブラチ ンやマイ ト マイ シ ンじの ような抗腫瘍剤は、 フ リ ーラ ジカルを産生することにより、 腫癘細 胞に細胞毒性を及ぼす (Oyanagi, Y. ら、 Biochem. Pharmacol., 26 , 473-476, 1997 ; Nakano, H. ら、 Biochem. Biop ys. Acta. , 796, 285-293, 1984)が、 IL— 6がフ リ ーラ ジカル分解作用を有するマン ガネーススーパーォキサイ ドジスムテース (Manganese superoxide dismutase ; MnSOD) の発現を促進し、 1L— 6抗体が促進された Mn SOD 発現を抑制することが知られている (Ono, M. , ら、 Biochem. Bi ophys. Res. Commun. , 182, 1100 - 1107, 1992 ; Dougal 1, W. C. ら、 Endocrinology, 129, 2376-2384, 1991)0 Platinum compounds such as cis-bratin and antitumor agents such as mitomycin C induce apoptosis in tumor cells, whereas IL-16 suppresses apoptosis induced by antitumour agents (Kerr, J. et al., Cancer 73, 2013-2026, 1994; Sachs, L. et al., Blood 82, 15-21, 1993). In addition, antitumor agents such as cisplatin and mitomycin produce cytotoxicity in tumor-producing cells by producing free radicals (Oyanagi, Y. et al., Biochem. Pharmacol. Nakano, H. et al., Biochem. Biophys. Acta., 796, 285-293, 1984) reported that IL-6 has a free radical-degrading activity. It is known that the expression of manganese superoxide dismutase (MnSOD) is promoted, and that the 1L-6 antibody suppresses the promoted expression of Mn SOD (Ono, M., et al., Biochem. Biophys. Res. Commun., 182, 1100-1107, 1992; Dougal 1, WC et al., Endocrinology, 129, 2376-2384, 1991) 0
しかしながら、 これらの報告は、 IL一 6の生物学的活性を遮断す ることにより抗腫瘼剤の作用を增強させることについて述べたもの はなく、 また、 実際に抗腫瘍剤の作用増強剤と して IL一 6 アンタゴ ニス 卜の使用を試みたものはなかった。  However, none of these reports mentions enhancing the action of antitumor agents by blocking the biological activity of IL-16. None attempted to use IL-16 Antagonist.
これまで腫瘍の治療には、 抗腫瘻剤が使用されていたが、 これら を多量に使用すると吐き気 · 嘔吐、 肾、 肝機能障害や骨髄機能抑制 といった好ま しく ない副作用が生ずることから、 腫瘍細胞に対し、 抗腫瘍作用を十分に発揮するような必要量を投与することが危険な 場合もあった。 また、 通常の抗腫瘍剤を用いる化学療法では効果が みられない治療抵抗性の腫瘍があり、 このような腫瘍の抗腫瘍剤に 対する感受性を上昇させる作用増強剤の登場が待たれていた。 本発明の目的は、 抗腫瘍剤の作用を補助、 増強し、 抗腫瘍剤に治 療抵抗性の腫瘍細胞の感受性を上昇させるような新しい抗腫瘍剤の 作用増強剤を提供することである。 より詳しく は、 本発明は I L一 6 ア ンタゴニス トを含んでなる、 抗腫瘍剤の作用増強剤を提供する。 より詳しく は、 本発明は I L一 6 アンタゴニス トを含んでなる、 抗腫 瘍作用を有する化学療法剤の作用増強剤を提供する。 発明の開示 Until now, antitumor agents have been used to treat tumors.However, if these drugs are used in large amounts, undesired side effects such as nausea and vomiting, 肾, hepatic dysfunction and bone marrow dysfunction may occur. On the other hand, it was sometimes dangerous to administer the required amount to sufficiently exert the antitumor effect. In addition, there are tumors resistant to treatment that are ineffective with conventional chemotherapy using antitumor agents, and the emergence of an action enhancer that increases the sensitivity of such tumors to antitumor agents has been awaited. An object of the present invention is to provide a new antitumor agent action enhancer that assists and enhances the action of an antitumor agent and increases the sensitivity of tumor cells resistant to treatment with the antitumor agent. More specifically, the present invention provides an antitumor agent action enhancer comprising IL-16 antagonist. More specifically, the present invention provides an action enhancer for a chemotherapeutic agent having an antitumor effect, comprising an IL-16 antagonist. Disclosure of the invention
本発明者らは、 腫瘍細胞の抗腫瘍剤に対する感受性の変化に I L一 6 アンタゴニス トが及ぼす影響について鋭意検討を重ねた結果、 I L ― 6抗体または I L一 6R抗体などの I L— 6 ア ンタゴニス トが腫瘍細胞 の抗腫瘍剤に対する感受性を上昇させ、 より低用量の抗腫瘍剤で治 療効果が認められること、 さらには、 通常の抗腫瘍剤には治療抵抗 性を示す腫瘍に対し I L— 6 ア ンタゴニス トを含んでなる作用増強剤 を併用するこ とにより治療効果が現れることを見出し、 本発明を完 成した o  The present inventors have conducted intensive studies on the effects of IL-16 antagonists on changes in the sensitivity of tumor cells to antitumor agents, and have found that IL-6 antagonists such as IL-6 antibody or IL-16R antibody. Increased the sensitivity of tumor cells to the antitumor agent, and a lower dose of the antitumor agent showed a therapeutic effect. The present inventors have found that a therapeutic effect can be exhibited by using an action enhancer containing an antagonist in combination, and have completed the present invention.o
すなわち、 本発明は I L一 6 アンタゴニス トを含んでなる、 抗腫瘍剤 の作用増強剤に関する。 より詳しく は、 本発明は I L一 6 ア ンタゴニ ス トを含んでなる、 抗腫瘍作用を有する化学療法剤の作用増強剤に 関する。 That is, the present invention relates to an antitumor agent action enhancer comprising IL-16 antagonist. More specifically, the present invention relates to an agent for enhancing the action of a chemotherapeutic agent having an antitumor effect, which comprises an IL-16 antagonist.
I L— 6 アンタゴニス トと しては、 I L— 6 に対する抗体、 —6Rに 対する抗体等が好ま し く 、 例えばこれらのモノ ク ローナル抗体が好 ま しい。 具体的なモノ ク ローナル抗体と しては PM— 1 抗体、 又はヒ ト型化 PM— 1 抗体が挙げられる。 また、 I L一 6 ア ンタゴニス 卜と組 合わせて使用される抗腫瘍剤と しては、 化学療法剤、 例えば抗腫瘍 活性を有する白金化合物、 マイ トマイ シ ン C等が挙げられる。 白金 化合物と しては、 シスブラチン、 カルボブラチン、 254- S, DWA- 21 1 4R, NK- 121等が挙げられる。 図面の簡単な説明 As the IL-6 antagonist, an antibody against IL-6, an antibody against -6R and the like are preferable, and for example, these monoclonal antibodies are preferable. Specific examples of the monoclonal antibody include a PM-1 antibody and a humanized PM-1 antibody. Examples of the antitumor agent used in combination with the IL-16 antagonist include chemotherapeutic agents, for example, a platinum compound having antitumor activity, mitomycin C, and the like. Platinum compounds include cisplatin, carboplatin, 254-S, DWA-21 1 4R, NK-121 and the like. BRIEF DESCRIPTION OF THE FIGURES
図 1 A及び図 I Bは、 0, 0.1, 1 または 10〃 g Zmlの濃度のシ スプラチンと IL— 6抗体 (図 1 A) または IL— 6R抗体 (図 1 B) が 共存したときの腎細胞癌株 Caki— 1 に対する細胞傷害活性を示す。 ♦は、 シスプラチンのみ、 圈は、 シスプラチンと 0.1〃 g Zmlの IL - 6抗体または IL一 6R抗体の共存下、 ▲は、 シスブラチンと l 〃 g ノ mlの IL— 6抗体または IL— 6R抗体の共存下、 参は、 シスブラチン と 10 /1111の 1しー 6抗体または IL—6R抗体の共存下における細胞 傷害活性 (%) を示す。 Figures 1A and IB show kidney cells in the presence of 0, 0.1, 1 or 10 µg Zml of cisplatin and IL-6 antibody (Figure 1A) or IL-6R antibody (Figure 1B). Shows cytotoxic activity against cancer strain Caki-1. ♦ indicates only cisplatin, circle indicates coexistence of cisplatin and 0.1 と g Zml of IL-6 antibody or IL-16R antibody, indicates cisplatin and l〃g noml of IL-6 antibody or IL-6R antibody presence, ginseng represents the cytotoxic activity (%) in the presence of 1 Sea 6 antibody or IL-6R antibody of Shisuburachin and 10/1111.
図 2 A及び図 2 Bは、 0, 0.1, 1 または 10 g Zmlの濃度のマ ィ トマイ シン Cと IL— 6抗体 (図 2 A) または IL— 6R抗体 (図 2 B ) が共存したときの腎細胞癌株 Caki— 1 に対する細胞傷害活性を示 す。 ♦は、 マイ トマイ シン Cのみ、 園は、 マイ トマイ シン Cと 0.1 gZmlの IL一 6抗体または IL— 6R抗体の共存下、 ▲は、 マイ トマ イ シンじと 1 g/mlの IL一 6抗体または IL— 6R抗体の共存下、 參 は、 マイ トマイ シン Cと 10// g Zmlの IL— 6抗体または IL一 6R抗体 の共存下における細胞傷害活性 ( ) を示す。  Figures 2A and 2B show the results when mitomycin C and IL-6 antibody (Figure 2A) or IL-6R antibody (Figure 2B) coexist at a concentration of 0, 0.1, 1 or 10 g Zml. Shows cytotoxic activity against renal cell carcinoma cell line Caki-1. ♦ indicates mitomycin C only, garden shows mitomycin C in the presence of 0.1 gZml IL-16 antibody or IL-6R antibody. ▲ indicates mitomycin C and 1 g / ml IL-16 In the presence of the antibody or the IL-6R antibody, the reference shows the cytotoxic activity () in the presence of the mitomycin C and 10 // g Zml of the IL-6 antibody or the IL-16R antibody.
図 3 A及び図 3 Bは、 0 , 0.1, 1 または 10 g /mlの濃度のシ スプラチンと IL— 6抗体 (図 3 A) または 1L一 6R抗体 (図 3 B ) が 共存したときの腎細胞癌株 Caki— 1 /DDP に対する細胞傷害活性を 示す。 ♦は、 シスブラチンのみ、 酾は、 シスブラチンと 0. l〃 g mlの IL— 6抗体または IL_ 6R抗体の共存下、 ▲は、 シスブラチンと 1 〃 g/mlの IL— 6抗体または 1L一 6R抗体の共存下、 眷は、 シスプ ラチンと 10 g/mlの IL— 6抗体または 1L一 6R抗体の共存下におけ る細胞傷害活性 (%) を示す。 図 4 A及び図 4 Bは、 0, 0.1, 1 または 10 z g Zmlの濃度のシ スブラチンと IL— 6抗体 (図 4 A) または IL— 6R抗体 (図 4 B ) が 共存したときの腎細胞癌株 ACHNに対する細胞傷害活性を示す。 ♦は 、 シスプラ チ ンのみ、 丽は、 シスプラチ ンと 0.1〃 g Zmlの IL— 6 抗体または IL一 6R抗体の共存下、 ▲は、 シスブラチンと FIGS. 3A and 3B show the results of the renal development in the presence of 0, 0.1, 1 or 10 g / ml of cisplatin and IL-6 antibody (FIG. 3A) or 1L-16R antibody (FIG. 3B). Shows cytotoxic activity against cell carcinoma cell line Caki-1 / DDP. ♦ indicates only cisplatin, 酾 indicates coexistence of cisplatin and 0.1 μg ml of IL-6 antibody or IL_6R antibody, and ▲ indicates cisplatin and 1 μg / ml IL-6 antibody or 1L-16R antibody Indicates the cytotoxic activity (%) in the co-presence of cisplatin and 10 g / ml of the IL-6 antibody or 1L-16R antibody. Figures 4A and 4B show renal cells in the presence of 0, 0.1, 1 or 10 zg Zml of cisplatin and IL-6 antibody (Figure 4A) or IL-6R antibody (Figure 4B). Fig. 4 shows cytotoxic activity against cancer cell line ACHN. ♦ indicates only cisplatin, 丽 indicates coexistence of cisplatin and 0.1 〃g Zml of IL-6 antibody or IL-16R antibody, ▲ indicates cisbratin
の IL一 6抗体または — 6R抗体の共存下、 譬は、 シスブラチンと 10 gZmlの IL— 6抗体または IL—6R抗体の共存下における細胞傷害 活性 (%) を示す。  In the co-presence of the IL-16 or IL-6R antibody, the pareto shows the cytotoxic activity (%) in the co-presence of cisplatin and 10 gZml of the IL-6 or IL-6R antibody.
図 5 A及び図 5 Bは、 0, 0.1, 1 または 10/ g Zmlの濃度のシ スプラチンと IL— 6抗体 (図 5 A) または IL—6R抗体 (図 5 B ) が 共存したときの腎細胞癌株 A704に対する細胞傷害活性を示す。 ♦は 、 シスプラチンのみ、 画は、 シスプラチンと 0. lz g/mlの IL— 6 抗体または IL— 6R抗体の共存下、 ▲は、 シスブラチンと 1 gZml の IL— 6抗体または 1L一 6R抗体の共存下、 秦は、 シスブラチンと 10 g Zmlの IL一 6抗体または 一 6R抗体の共存下における細胞傷害 活性 (%) を示す。  FIGS. 5A and 5B show the results of the renal development when cisplatin and IL-6 antibody (FIG. 5A) or IL-6R antibody (FIG. 5B) coexist at concentrations of 0, 0.1, 1 or 10 / g Zml. 3 shows cytotoxic activity against cell carcinoma line A704. ♦: only cisplatin, fraction: coexistence of cisplatin and 0.1 lz g / ml of IL-6 antibody or IL-6R antibody, ▲: coexistence of cisplatin and 1 gZml of IL-6 antibody or 1L-16R antibody The lower column shows the cytotoxic activity (%) in the presence of cisplatin and 10 g Zml of IL-16 antibody or 16R antibody.
図 6 A及び図 6 Bは、 0 , 0.1, 1 または 10 g /mlの濃度のシ スプラチンと 1L— 6抗体 (図 6 A) または 一 6R抗体 (図 6 B ) 力く 共存したときの患者 1 から得られた新鮮腎細胞癌に対する細胞傷害 活性を示す。 ♦は、 シスブラチンのみ、 顬は、 シスブラチンと 0.1 g Zmlの IL— 6抗体または IL— 6R抗体の共存下、 ▲は、 シスブラ チンと 1 / g Zmlの IL— 6抗体または IL— 6R抗体の共存下、 秦は、 シスブラチンと 10/ gZmlの IL— 6抗体または 1L—6R抗体の共存下 における細胞傷害活性 (%) を示す。 .  Figures 6A and 6B show patients with vigorous coexistence of cisplatin at 0, 0.1, 1 or 10 g / ml with 1L-6 antibody (Figure 6A) or 16R antibody (Figure 6B). 1 shows the cytotoxic activity against fresh renal cell carcinoma obtained from FIG. ♦ indicates only cisplatin, 顬 indicates coexistence of 0.1 g Zml of IL-6 antibody or IL-6R antibody, and ▲ indicates coexistence of 1 / g Zml of IL-6 antibody or IL-6R antibody. The lower column shows cytotoxic activity (%) in the presence of cisplatin and 10 / gZml of IL-6 antibody or 1L-6R antibody. .
図 7 A及び図 7 Bは、 0, 0.1, 1 または 10〃 g Zmlの濃度のシ スプラチンと IL— 6抗体 (図 7 A) または IL— 6R抗体 (図 7 B ) 力く 共存したときの患者 2 から得られた新鮮腎細胞癌に対する細胞傷害 活性を示す。 ♦は、 シスブラチンのみ、 疆は、 シスブラチンと 0.1 gZm】の】 L— 6抗体または IL— 6R抗体の共存下、 ▲は、 シスブラ チンと 1 a gZmlの IL一 6抗体または IL— 6R抗体の共存下、 翁は、 シスブラチンと 10 / g mlの IL— 6抗体または IL— 6R抗体の共存下 における細胞傷害活性 (%) を示す。 Figures 7A and 7B show the results when 0, 0.1, 1 or 10 µg Zml of cisplatin and IL-6 antibody (Fig. 7A) or IL-6R antibody (Fig. 7B) coexist vigorously. Cytotoxicity against fresh renal cell carcinoma obtained from patient 2 Show activity. ♦ indicates the presence of only cisplatin, and Xin indicates the presence of 0.1 gZm of cisplatin; L-6 antibody or IL-6R antibody. ▲ indicates the presence of 1 agZml of IL-16 antibody or IL-6R antibody of cisplatin. In the figure, Okina shows the cytotoxic activity (%) in the presence of cisplatin and 10 / g ml of IL-6 antibody or IL-6R antibody.
図 8 A及び図 8 Bは、 0, 0.1, 1 または 10〃 g Zmlの濃度のシ スブラチンと IL一 6抗体 (図 8 A) または IL— 6R抗体 (図 8 B ) が 共存したときの患者 3から得られた新鮮腎細胞癌に対する細胞傷害 活性を示す。 ♦は、 シスブラチンのみ、 鼴は、 シスブラチンと 0.1 gノ mlの IL— 6抗体または IL— 6R抗体の共存下、 ▲は、 シスブラ チンと 1 /z g Zmlの IL一 6抗体または IL一 6R抗体の共存下、 秦は、 シスブラチンと 10 g /mlの IL一 6抗体または IL— 6R抗体の共存下 における細胞傷害活性 (%) を示す。  Figures 8A and 8B show patients with coexisting 0, 0.1, 1 or 10 µg Zml of cisplatin and IL-16 antibody (Figure 8A) or IL-6R antibody (Figure 8B). 3 shows the cytotoxic activity against fresh renal cell carcinoma obtained from 3. ♦ indicates only cisplatin, 鼴 indicates coexistence of cisplatin and 0.1 g of IL-6 antibody or IL-6R antibody, and ▲ indicates that of cisplatin and 1 / zg Zml of IL-16 antibody or IL-16R antibody. Hata shows the cytotoxic activity (%) in the presence of cisplatin and 10 g / ml of IL-16 antibody or IL-6R antibody.
図 9 A及び図 9 Bは、 0 , 1 , 10または 100 g Zmlの濃度の力 ルポプラチンと — 6抗体 (図 9 A) または IL—6R抗体 (図 9 B ) が共存したときの肾細胞癌株 Caki— 1 に対する細胞傷害活性を示す 。 ♦は、 カルボブラチンのみ、 國は、 カルボブラチンと 0. l g Z mlの Iし— 6抗体または IL— 6R抗体の共存下、 ▲は、 カルボブラチン と 1 u g /mlの — 6抗体または IL— 6R抗体の共存下、 秦は、 カル ポプラチンと 10 8 11)1の 1しー 6抗体または IL一 6R抗体の共存下に おける細胞傷害活性 (%) を示す。  FIGS. 9A and 9B show 肾 cell carcinoma in the presence of lipoplatin at 0, 1, 10 or 100 g Zml at the concentration of —6 antibody (FIG. 9A) or IL-6R antibody (FIG. 9B). Shows cytotoxic activity against strain Caki-1. ♦ indicates only carbobratin, and Japan indicates the presence of 0.1 lg Zml of I-6 antibody or IL-6R antibody in the country. ▲ indicates 1 ug / ml of 6 antibody or IL-6R antibody with carboplatin. In the coexistence, Hata shows the cytotoxic activity (%) in the coexistence of calpoplatin and 10 8 11) 1 1-6 antibody or IL-16R antibody.
図 10中の Aは、 培養液 (コ ン トロール) 、 10iz g Zmlのシスブラ チ ン、 10/ g Zmlの IL一 6抗体または IL一 6R抗体で処理した腎細胞 癌株 Caki _ 1 において GST— 7Γ mRNA 発現を調べるための GST— ττ cDNA プローブを用いた Caki— 1 の全 RNA のノザンブロ ッ トの結果 を示す図である。 レー ン 1 は、 培養液のみ、 レー ン 2 はシスプラチ ン、 レーン 3 は IL— 6抗体、 レーン 4 は 1し一 6R抗体で処理している 。 図 10中の Bは、 ェチジゥムブ口マイ ド染色を施した、 ノザンプロ ッ 卜に用いたゲルの図である。 各レーンには RNA が存在することを 示す。 具体的な説明 A in FIG. 10 indicates GST-in renal cell carcinoma cell line Caki_1 treated with culture medium (control), 10iz g Zml of cis-bratin, 10 / g Zml of IL-16 antibody or IL-16R antibody. FIG. 7 shows the results of Northern blotting of total Caki-1 RNA using a GST-ττ cDNA probe for examining 7Γ mRNA expression. Lane 1 was treated with culture medium only, lane 2 was treated with cisplatin, lane 3 was treated with IL-6 antibody, and lane 4 was treated with 1-6R antibody . B in FIG. 10 is a diagram of a gel used for Northern plots, which was stained with ethidium umide. Each lane indicates the presence of RNA. Specific description
本発明の 1 L— 6 アンタゴニス トを含んでなる抗腫瘍剤の作用増強 剤とは、 腫瘍を治療するに際し、 抗腫瘍剤と併用されることで、 抗 腫瘍作用を增強する。 また、 抗腫瘍剤の必要用量を低減させ、 さ ら には通常の化学療法では治療効果が認められない治療抵抗性の腫瘍 に対しても、 抗腫瘍剤の感受性を上昇させる効果を有する。  The antitumor agent-enhancing agent comprising 1L-6 antagonist of the present invention enhances the antitumor effect when used in combination with an antitumor agent in treating a tumor. In addition, it has the effect of reducing the required dose of an antitumor agent and increasing the sensitivity of the antitumor agent even to a treatment-resistant tumor for which a therapeutic effect is not recognized by ordinary chemotherapy.
本発明の作用増強剤により抗腫瘍作用が増強される抗腿瘍剤は、 腫瘍細胞に作用 して腫瘍細胞の発育と増殖を抑制し、 腫瘍の治療効 果を有する化学療法剤である。 化学療法剤には、 アルキル化剤、 代 謝拮抗剤、 抗腫瘍抗生剤、 植物由来アル力ロイ ド、 ホルモ ン療法剤 、 白金化合物などがある。 このような抗腫瘍剤の中でも、 抗腫瘍抗 生剤であるマイ トマイ シ ン Cゃ抗腫瘍作用を有する白金化合物が特 に好ま しい。 白金化合物は白金原子を有し、 他の原子と錯体を形成 している。 このものは、 DNA と結合することにより、 腫瘍細胞の DN A 合成を阻害し、 腫瘍細胞の分裂を阻害して抗腫癟作用を示す。 こ れまでに抗腫瘍作用を有する白金化合物と して、 シスブラチ ン (ci s-d i ammi ne d i ch 1 orop 1 a t i num (11)、 下言己の構造式を有する) 、  The anti-tumor agent whose antitumor effect is enhanced by the action enhancer of the present invention is a chemotherapeutic agent that acts on tumor cells to suppress the growth and proliferation of tumor cells and has a therapeutic effect on tumors. Chemotherapeutic agents include alkylating agents, metabolic antagonists, antitumor antibiotics, plant-derived alkyloids, formone therapeutic agents, and platinum compounds. Among such antitumor agents, a platinum compound having an antitumor effect of mitomycin C ゃ, which is an antitumor agent, is particularly preferred. Platinum compounds have a platinum atom and form a complex with other atoms. By binding to DNA, it inhibits the synthesis of DNA in tumor cells, inhibits the division of tumor cells, and has an antitumor effect. To date, platinum compounds having antitumor activity include cisplatin (cis-diamminedich1 orop1 atinum (11), having the following structural formula).
NH,、 ノ ,C1 NH,ノ 、C1 カノレホブラチ ン ( c i s-d i ammi ne (1, 1 -cycl obu taned i carboxy 1 a t o; - Platinum (II) 、 下記の構造式を有する) 、 NH, NO, C1 NH, NO, C1 canolehobratin (cisd i ammine (1, 1-cycl obu taned i carboxy 1 ato;-Platinum (II), having the following structural formula),
Figure imgf000012_0001
Figure imgf000012_0001
254-S ((Glycolato-0, 0' ) d i ammi nep 1 a t i num 、 下記の構造式 を有する) 、 254-S ((Glycolato-0, 0 ') d i ammi nep 1 a t i num, having the following structural formula),
Figure imgf000012_0002
Figure imgf000012_0002
DWA-2114R ((-) -(R) -2 -am inomethyl pyrrol idine (1, 1-cyclobutane di carboxylatoplat inum (11)、 下記の構造式を有する) 、  DWA-2114R ((-)-(R) -2 -am inomethyl pyrrol idine (1, 1-cyclobutane di carboxylatoplat inum (11), having the following structural formula),
Figure imgf000012_0003
Figure imgf000012_0003
NK-121 ((R)-ci s-2-methy 1 -1, 4-butane.diamine (1, 1-cyclobutaned icarboxylato) plat inum (I I) 、 下記の構造式を有する) 、
Figure imgf000013_0001
ォキサリブラチン (oxaliplatin ; USAN, Oxalato (trans-1, 2-cyc lohexanediamine) platinum (11)、 下 の構造式を有 _9 ) 、
NK-121 ((R) -cis-2-methy 1 -1,4-butane.diamine (1, 1-cyclobutaned icarboxylato) plat inum (II), having the following structural formula),
Figure imgf000013_0001
Okisariburachin (oxaliplatin; USAN, Oxalato (trans -1, 2-cyc lohexanediamine) platinum (11), have a structural formula below _ 9),
Figure imgf000013_0002
Figure imgf000013_0002
TRK-710 ((alpha-acetyl -gamma-methyl tetronate) z-( I -1, 2-diamin ocyclohexane) platinum (II) 、 下記の構造式を有する) TRK-710 ((alpha-acetyl-gamma-methyl tetronate) z- (I-1, 2-diaminocyclohexane) platinum (II), having the following structural formula)
Figure imgf000013_0003
等が知られている。 これらの白金化合物のう ちで特に、 上記シスプ ラチン、 カルポプラチン、 DWA-2114R が好ま し く 例示される。
Figure imgf000013_0003
Etc. are known. Among these platinum compounds, cisplatin, carboplatin and DWA-2114R are particularly preferred.
これらの抗腫瘍剤は、 常法により製剤化される。 例えば、 白金化 合物の場合、 必要ならば補助剤と と もに医薬と して用いる単体と混 合して、 経口的にまた、 非経口的に、 好ま し く は注射剤と して用い られる。 注射剤とする場合には、 蒸留水あるいは塩化ナ ト リ ウム、 塩化カ リ ウム等の塩溶液または、 ブ ドウ糖溶液、 生理食塩水に混和 するのがよい。 これらの製剤中の抗腫瘍剤の量は、 患者の年齢、 症 状等により、 使用に便利な単位量が望まれ、 例えば、 成人の腫瘍治 療に用いられる場合、 一日一回 10— 2000mgZm2 (体積面積) を投与 し、 投与量により 5 日間の連投あるいは投与間に 1 一 4週の休薬期 間を設けてもよい。 These antitumor agents are formulated in a usual manner. For example, in the case of a platinum compound, if necessary, it is mixed with an adjuvant and a simple substance used as a drug, and used orally or parenterally, preferably as an injection. Can be In the case of an injection, it is advisable to mix it with distilled water or a salt solution such as sodium chloride or potassium chloride, a glucose solution or a physiological saline solution. The amount of the antitumor agent in these preparations is desired to be a convenient unit dose depending on the age and condition of the patient. For example, when used for the treatment of tumors in adults, 10 to 2000 mgZm once a day 2 (volume area) may be administered, and depending on the dose, continuous administration for 5 days or a drug holiday of 114 weeks between administrations may be provided.
本発明の作用増強剤により抗腫瘍作用が認められる腫瘍細胞は、 IL— 6Rを有し、 IL— 6 を一つの生理活性物質と して増殖および Zま たは治療抵抗性を示す腫瘍である。 このような腫瘍と しては、 腎細 胞癌 (Miki, S. ら、 FEBS Letter, 250, 607-610, 1989)、 骨髄腫 ( Kawano, M. ら、 Nature, 332, 83-85, 1988) 、 卵巣ガン (Kobayash i, H. ら、 第 53回日本がん学会総会講演要旨集 271 頁、 874, 1994) 、 EBウィルス感染 B リ ンパ腫 (Tosata, G. ら、 J. Virol. , 64, 3033 -3041, 1990)、 成人 T細胞白血病 (Sawada, T. k Takatsuki, K. Br . J. Cancer, 62, 923-924, 1990) 、 前立腺がん (Siegall, C. B. ら 、 Cancer Res. , 50, 7786-7788, 1990) 、 Kaposi肉腫 (Miles, S. A . ら、 Proc. Natl. Acad. Sci. 87, 4068-4072, 1990)等が知られてい る σ  Tumor cells having an antitumor effect by the action enhancer of the present invention are tumors having IL-6R and exhibiting proliferation and Z or treatment resistance using IL-6 as one physiologically active substance. . Such tumors include renal cell carcinoma (Miki, S. et al., FEBS Letter, 250, 607-610, 1989) and myeloma (Kawano, M. et al., Nature, 332, 83-85, 1988). ), Ovarian cancer (Kobayash i, H. et al., Proceedings of the 53rd Annual Meeting of the Japanese Cancer Society, 271 pages, 874, 1994), EB virus-infected B lymphoma (Tosata, G. et al., J. Virol. 64, 3033-3041, 1990), adult T-cell leukemia (Sawada, T. k Takatsuki, K. Br. J. Cancer, 62, 923-924, 1990), prostate cancer (Siegall, CB et al., Cancer Res. , 50, 7786-7788, 1990) and Kaposi sarcoma (Miles, S.A. et al., Proc. Natl. Acad. Sci. 87, 4068-4072, 1990).
本発明で使用される IL_ 6 アンタゴニス トは、 IL— 6 による シグ ナル伝達を遮断し、 1L一 6 の生物学的活性を阻害する ものであれば 、 その由来を問わない。 IL一 6 アンタゴニス トと しては、 IL— 6抗 体、 IL— 6R抗体、 gpl30 抗体、 IL— 6改変体、 IL— 6Rのア ンチセン スオ リ ゴヌ ク レオチ ドあるいは IL一 6 または IL— 6Rの部分べプチ ド 等が挙げられる。 The IL_6 antagonist used in the present invention may be of any origin as long as it blocks signal transmission by IL-6 and inhibits the biological activity of 1L-16. As an IL-6 antagonist, IL-6 Body, IL-6R antibody, gpl30 antibody, IL-6 variant, antisense oligonucleotide of IL-6R, partial peptide of IL-16 or IL-6R, and the like.
本発明で 1L一 6 アンタゴニス ト と して使用される抗体、 たとえば 、 IL— 6抗体、 IL— 6R抗体、 あるいは gpl30 抗体はその由来および 種類 (モノ ク ローナル、 ポリ ク ローナル) を問わないが、 特に哺乳 動物由来のモノ ク ローナル抗体が好ま しい。 これら抗体は IL— 6, IL— 6Rあるいは gpl30 と結合することにより、 IL— 6 と IL—6Rまた は IL— 6Rと gpl30 の結合を阻害して 1L_ 6 のシグナル伝達を遮断し 、 1L一 6 の生物学的活性を阻害する抗体である。  Antibodies used as 1L-16 antagonists in the present invention, for example, IL-6 antibody, IL-6R antibody, or gpl30 antibody may be of any origin and type (monoclonal, polyclonal). In particular, a monoclonal antibody derived from a mammal is preferred. By binding to IL-6, IL-6R or gpl30, these antibodies inhibit the binding of IL-6 to IL-6R or IL-6R and gpl30, block 1L_6 signaling, and Is an antibody that inhibits the biological activity of
モノ ク ローナル抗体の産生細胞の動物種は哺乳類であれば特に制 限されず、 ヒ 卜抗体またはヒ ト以外の哺乳動物由来であってよい。 ヒ ト以外の哺乳動物由来のモノ ク ローナル抗体と しては、 その作成 の簡便さからゥサギあるいはげつ歯類由来のモノ ク ローナル抗体が 好ま しい。 げっ歯類と しては、 特に制限されないが、 マウス、 ラ ッ ト、 ハムスターなどが好ま し く例示される。  The animal species of the cell producing the monoclonal antibody is not particularly limited as long as it is a mammal, and may be derived from a human antibody or a mammal other than human. As a monoclonal antibody derived from a mammal other than human, a monoclonal antibody derived from a rabbit or a rodent is preferred because of its ease of preparation. The rodent is not particularly limited, but mouse, rat, hamster and the like are preferably exemplified.
このよ う な抗体は、 IL— 6抗体と しては、 MH166 抗体 (Matsuda ら、 Eur. J. Immunol. 18 : 951-956, 1988)や SK2抗体 (Satoら、 第 21回日本免疫学会総会、 学術記録、 21 : 116, 1991) 等が挙げられ る。 IL— 6R抗体と しては、 PM— 1 抗体 (Hirataら、 J. Immunol. 143 : 2900-2906, 1989) 、 AUK12- 20抗体、 AUK64- 7 抗体あるいは AUK1 46-15 抗体 (国際特許出願公開番号 W092— 19759)などが挙げられる このような抗体のう ち、 特に IL— 6R抗体が好ま し く 、 その具体例 と しては上記 PM- 1 抗体が挙げられる。  Such antibodies include MH166 antibody (Matsuda et al., Eur. J. Immunol. 18: 951-956, 1988) and SK2 antibody (Sato et al., 21st Annual Meeting of the Immunological Society of Japan). Scientific Records, 21: 116, 1991). Examples of the IL-6R antibody include PM-1 antibody (Hirata et al., J. Immunol. 143: 2900-2906, 1989), AUK12-20 antibody, AUK64-7 antibody, and AUK146-5-15 antibody (International Patent Application Publication). Among them, the IL-6R antibody is particularly preferable, and specific examples thereof include the above-mentioned PM-1 antibody.
モノ ク ロ ーナル抗体は、 基本的には公知技術を使用 し、 以下のよ うにして作成できる。 すなわち、 IL— 6 , IL— 6Rあるいは gpl30 を 感作抗原と して使用 して、 これを通常の免疫方法に したがって免疫 し、 得られる免疫細胞を通常の細胞融合法によつて公知の親細胞と 融合させ、 通常のスク リ ーニング法により、 モノ ク ローナルな抗体 産生細胞をスク リ ーニングするこ とによって作成できる。 A monoclonal antibody can be basically produced using a known technique as follows. That is, IL-6, IL-6R or gpl30 It is used as a sensitizing antigen, and is immunized according to a conventional immunization method, and the obtained immune cells are fused with a known parent cell by a normal cell fusion method, and are then subjected to a normal screening method. It can be prepared by screening monoclonal antibody-producing cells.
より具体的には、 モノ ク ローナル抗体を作成するには次のよう に すればよい。 例えば、 前記感作抗原と してはヒ ト由来のものが好ま し く 、 ヒ ト IL— 6の場合、 Hiranoら、 Nature, 324 : 73, 1986に開 示されたヒ ト IL一 6 の遗伝子配列を用いるこ とによって得られる。 ヒ ト ー 6の遺伝子配列を公知の発現ベク ター系に挿人して適当な 宿主細胞を形質転換させた後、 その宿主細胞中または、 培養上清中 から目的の IL— 6 タ ンパク質を精製し、 この精製 IL— 6 タ ンパク質 を感作抗原と して用いればよい。  More specifically, a monoclonal antibody can be prepared as follows. For example, the sensitizing antigen is preferably a human-derived antigen. In the case of human IL-6, the human sensitizing antigen is a human IL-16 which is disclosed in Hirano et al., Nature, 324: 73, 1986. It is obtained by using gene sequences. After the human 6 gene sequence is inserted into a known expression vector system to transform an appropriate host cell, the desired IL-6 protein is isolated from the host cell or the culture supernatant. After purification, the purified IL-6 protein may be used as a sensitizing antigen.
ヒ ト IL一 6Rの場合、 欧州特許出願公開番号 EP325474号に開示され た遺伝子配列を用いて上記ヒ 卜 IL一 6 と同様の方法に従えば IL— 6R タンパク質を得るこ とができる。 IL— 6Rは細胞膜上に発現している ものと細胞膜より離脱している可溶性のもの(sIL— 6R) (Yasukawa ら、 J. Biochem. , 108, 673-676, 1990) との二種類がある。 sIL— 6Rは細胞膜に結合している IL一 6Rの主に細胞外領域から構成されて おり、 細胞膜貫通領域あるいは細胞膜貫通領域と細胞内領域が欠損 している点で膜結合型 IL一 6Rと異なっている。  In the case of human IL-16R, an IL-6R protein can be obtained by using the gene sequence disclosed in European Patent Application Publication No. EP325474 and following the same method as for human IL-16. There are two types of IL-6R: those expressed on the cell membrane and those soluble off the cell membrane (sIL-6R) (Yasukawa et al., J. Biochem., 108, 673-676, 1990). . sIL-6R is composed mainly of the extracellular region of IL-16R bound to the cell membrane, and differs from membrane-bound IL-16R in that the transmembrane region or the transmembrane region and the intracellular region are defective. Is different.
ヒ ト gpl30 の場合、 欧州特許出願公開番号 EP411946に開示されて いる遺伝子配列を用いて上記 IL— 6 と同様の方法に従えば、 gpl30 夕 ンパク質を得るこ とができる。  In the case of human gpl30, gpl30 protein can be obtained by using the gene sequence disclosed in European Patent Application Publication No. EP411946 and following the same method as for IL-6.
感作抗原で免疫される哺乳動物と しては、 特に限定される もので はないが、 細胞融合に使用する親細胞との適合性を考慮して選択す るのが好ま し く 、 一般的にはマウス、 ラ ッ ト、 ハムスター、 ゥサギ 等が使用される。 感作抗原を動物に免疫するには、 公知の方法にしたがって行われ る。 例えば、 一般的方法と して、 感作抗原を哺乳動物に腹腔内また は、 皮下に注射することにより行われる。 具体的には、 感作抗原をThe mammal to be immunized with the sensitizing antigen is not particularly limited, but is preferably selected in consideration of compatibility with the parent cell used for cell fusion. For this, mice, rats, hamsters, egrets, etc. are used. Immunization of an animal with a sensitizing antigen is performed according to a known method. For example, as a general method, a sensitizing antigen is injected into a mammal intraperitoneally or subcutaneously. Specifically, sensitizing antigen
PBS (Phosphate-Buffered Saline) や生理食塩水等で適当量に希釈 、 懸濁したものを所望により通常のア ジュバン ト、 例えば、 フ ロイ ン ト完全アジュバン トを適量混合し、 乳化後、 哺乳動物に 4 一 21日 毎に数回投与するのが好ま しい。 また、 感作抗原免疫時に適当な担 体を使用することができる。 After diluting and suspending an appropriate amount with PBS (Phosphate-Buffered Saline) or physiological saline, etc., add an appropriate amount of a normal adjuvant, for example, complete Freund's adjuvant, and emulsify the suspension. It is preferred to administer several doses every 4 to 21 days. In addition, an appropriate carrier can be used at the time of immunization with the sensitizing antigen.
このよ うに免疫し、 血清中に所望の抗体レベルが上昇するのを確 認した後に、 哺乳動物から免疫細胞が取り出され、 細胞融合に付さ れるが、 好ま しい免疫細胞と しては、 特に脾細胞が挙げられる。 前記免疫細胞と融合される他方の親細胞と しての哺乳動物のミエ ローマ細胞は、 すでに、 公知の種々の細胞株、 例えば、 P3 (P3x63A g8. 653) (J. Immunol. 123 : 1548, 1978), p3-Ul (Current Topic s in Micro-biology and Immunology 81 : 1-7, 1978), NS-1 (Eur . J. Immunol. 6 : 511-519, 1976), MPC-11 (Cell, 8 : 405-415, 1 976), SP2/0 (Nature, 276 : 269-270, 1978), F0 (J. Immunol. Met h. 35 : 1-21, 1980), S194 (J. Exp. Med. 148 : 313-323, 1978), R210 (Nature, 277 : 131-133, 1979)等が好適に使用される。  After immunization in this way and confirming that the level of the desired antibody in the serum is increased, immune cells are removed from the mammal and subjected to cell fusion. Preferred immune cells are particularly preferred. Splenocytes. Mammalian myeloma cells as the other parent cells fused with the immune cells are already known in various cell lines, for example, P3 (P3x63Ag8.653) (J. Immunol. 123: 1548, 1978), p3-Ul (Current Topics in Micro-biology and Immunology 81: 1-7, 1978), NS-1 (Eur. J. Immunol. 6: 511-519, 1976), MPC-11 (Cell, 8: 405-415, 1 976), SP2 / 0 (Nature, 276: 269-270, 1978), F0 (J. Immunol. Met h. 35: 1-21, 1980), S194 (J. Exp. Med) 148: 313-323, 1978), R210 (Nature, 277: 131-133, 1979) and the like are preferably used.
前記免疫細胞と ミエ口一マ細胞との細胞融合は基本的には公知の 方法、 たとえば、 ミ ルスティ ンらの方法 (Milsteinら、 Methods En zymol. 73 : 3-46, 1981) 等に準じて行う ことができる。  Cell fusion between the immune cells and myeoma cells is basically performed according to a known method, for example, the method of Milstein et al. (Milstein et al., Methods Enzymol. 73: 3-46, 1981). It can be carried out.
より具体的には、 前記細胞融合は例えば、 細胞融合促進剤の存在 下に通常の栄養培養液中で実施される。 融合促進剤と しては例えば 、 ポ リ エチレングリ コール (PEG)、 センダイウ ィ ルス (HVJ)等が使 用され、 更に所望により融合効率を高めるためにジメチルスルホキ シ ド等の補助剤を添加使用することもできる。 免疫細胞と ミ エローマ細胞との使用割合は、 例えば、 ミ エ口一マ 細胞に対して免疫細胞を 1 一 1 0倍とするのが好ま しい。 前記細胞融 合に用いる培養液と しては、 例えば、 前記ミ エローマ細胞株の増殖 に好適な RPM 1 1 640 培養液、 MEM 培養液、 その他、 この種の細胞培 養に用いられる通常の培養液が使用可能であり、 さ らに、 牛胎児血 清 (FC S )等の血清補液を併用するこ と もできる。 More specifically, the cell fusion is performed, for example, in a normal nutrient culture in the presence of a cell fusion promoter. As the fusion promoter, for example, polyethylene glycol (PEG), Sendai virus (HVJ) or the like is used, and if necessary, an auxiliary agent such as dimethyl sulfoxide is added to enhance the fusion efficiency. Can also be used. The ratio of the use of the immune cells to the myeloma cells is preferably, for example, 11 to 10 times the number of the immune cells to the myeoma cells. Examples of the culture medium used for cell fusion include RPM11640 culture medium and MEM culture medium suitable for the proliferation of the myeloma cell line, and other ordinary cultures used for cell culture of this type. A fluid can be used, and a serum replacement fluid such as fetal calf serum (FCS) can also be used in combination.
細胞融合は、 前記免疫細胞と ミ エローマ細胞との所定量を前記培 養液中でよ く混合し、 予め、 37 °C程度に加温した P EG 溶液、 例えば 、 平均分子量 1 000— 6000程度の P EG 溶液を通常、 30— 60 % ( w Z v ) の濃度で添加 し、 混合するこ とによって目的とする融合細胞 (ハ イブリ ドーマ) が形成される。 続いて、 適当な培養液を逐次添加し 、 遠心して上清を除去する操作を繰り返すことによりハイプリ ドー マの生育に好ま し く ない細胞融合剤等を除去できる。  For cell fusion, a predetermined amount of the immune cells and myeloma cells is mixed well in the culture medium, and a PEG solution previously heated to about 37 ° C., for example, an average molecular weight of about 1,000 to 6,000 The desired fused cells (hybridoma) are formed by adding and mixing the PEG solution of the above at a concentration of 30-60% (wZv). Subsequently, by repeating the operation of successively adding an appropriate culture solution and centrifuging to remove the supernatant, a cell fusion agent or the like unfavorable for the growth of the hybridoma can be removed.
当該ハイプリ ドーマは、 通常の選択培養液、 例えば、 HAT 培養液 (ヒポキサンチン、 ア ミ ノ ブテ リ ンおよびチ ミ ジンを含む培養液) で培養することにより選択される。 当該 HAT 培養液での培養は、 目 的とするハイプリ ドーマ以外の細胞 (非融合細胞) が死滅するのに 十分な時間、 通常数日〜数週間継続する。 ついで、 通常の限界希釈 法を実施し、 目的とする抗体を産生するハイプリ ドーマのスク リ ー 二ングおよび単一ク ロ一ン化が行われる。  The hybridoma is selected by culturing it in a normal selective culture solution, for example, a HAT culture solution (a culture solution containing hypoxanthine, aminobuterin and thymidine). Culture in the HAT culture medium is continued for a period of time sufficient to kill cells other than the target hybridoma (non-fused cells), usually several days to several weeks. Next, a conventional limiting dilution method is performed, and screening and monocloning of the hybridoma producing the desired antibody are performed.
このようにして作成されるモノ ク ローナル抗体を産生するハイブ リ ドーマは、 通常の培養液中で継代培養することが可能であり、 ま た、 液体窒素中で長期保存することが可能である。  The hybridoma producing the monoclonal antibody thus produced can be subcultured in a normal culture medium, and can be stored for a long time in liquid nitrogen. .
当該ハイプリ ドーマからモノ ク ローナル抗体を取得するには、 当 該ハイプリ ドーマを通常の方法にしたがい培養し、 その培養上清と して得る方法、 あるいはハイプリ ドーマをこれと適合性がある哺乳 動物に投与して増殖させ、 その腹水と して得る方法などが採用され る。 前者の方法は、 高純度の抗体を得るのに適しており、 一方、 後 者の方法は、 抗体の大量生産に適している。 To obtain a monoclonal antibody from the hybridoma, a method of culturing the hybridoma according to an ordinary method and obtaining the culture supernatant, or transferring the hybridoma to a mammal compatible therewith Injection, proliferation and ascites are used. You. The former method is suitable for obtaining high-purity antibodies, while the latter method is suitable for mass production of antibodies.
また、 モノ ク ローナル抗体は、 抗原を免疫して得られる抗体産生 細胞を細胞融合させて生ずるハイプリ ドーマから得られる ものだけ でなく 、 抗体遺伝子をク ローニングし、 適当なベク ターに組み込ん で、 これを公知の細胞株、 例えば、 COS, CH0等に導人し、 遺伝子組 換え技術を用いて産生させたモノ ク ローナル抗体を用いるこ とがで きる (例えば、 Vandamme, A- M. ら、 Eur. J. B i ochem., 192, 767-775 , 1990参照) 。  Monoclonal antibodies include not only those obtained from hybridomas produced by cell fusion of antibody-producing cells obtained by immunization with an antigen, but also those obtained by cloning an antibody gene and incorporating it into an appropriate vector. Can be used as a monoclonal antibody produced by using a gene recombination technique by introducing the gene into a known cell line, for example, COS, CH0, etc. (for example, Vandamme, A-M. Et al., Eur J. Biochem., 192, 767-775, 1990).
さ らに、 前記の方法により得られるモノ ク ローナル抗体は、 塩析 法ゲル濾過法、 ァフ ィ 二ティ ーク ロマ ト グラフ ィ 一法等の通常の精 製手段を利用 して高純度に精製することができる。  Furthermore, the monoclonal antibody obtained by the above-mentioned method can be purified to a high degree of purity using ordinary purification means such as a salting-out method, a gel filtration method, and an affinity chromatography method. It can be purified.
このようにして、 作成されるモノ ク ローナル抗体は、 放射免疫測 定法 (RIA)、 酵素免疫測定法 (E1A, ELISA) 、 蛍光抗体法 ( Immuno fluorescence Ana lysis)等の通常の免疫学的手段により抗原を高感 度かつ高精度で認識するこ とを確認することができる。  The monoclonal antibodies thus prepared can be obtained by ordinary immunological means such as radioimmunoassay (RIA), enzyme immunoassay (E1A, ELISA), and immunofluorescence assay (Immunofluorescence analysis). It can be confirmed that the antigen is recognized with high sensitivity and high accuracy.
本発明に使用されるモノ ク ローナル抗体は、 ハイプリ ドーマが産 生するモノ ク ローナル抗体に限られるものではなく 、 ヒ トに対する 異種抗原性を低下させるこ と等を目的と して人為的に改変したもの がより好ま しい。 例えば、 ヒ ト以外の哺乳動物、 例えば、 マウスの モソ ク ローナル抗体の可変領域と ヒ ト抗体の定常領域とからなるキ メ ラ抗体を使用するこ とができ、 このようなキメ ラ抗体は、 既知の キメ ラ抗体の製造方法、 特に遗伝子組換技法を用いて製造するこ と ができる。  The monoclonal antibody used in the present invention is not limited to the monoclonal antibody produced by the hybridoma, but may be artificially modified for the purpose of, for example, reducing the antigenicity to humans. What you do is better. For example, a chimeric antibody comprising a variable region of a mammalian non-human, eg, mouse, monoclonal antibody and a constant region of a human antibody can be used. It can be produced using known methods for producing chimeric antibodies, particularly using gene recombination techniques.
さ らに、 再構成 (reshaped) したヒ ト抗体を本発明に用いるこ と ができる。 これはヒ ト以外の哺乳動物、 たとえばマウス抗体の相補 性決定領域をヒ ト抗体の相補性決定領域へ移植したものであり、 そ の一般的な遺伝子組換手法も知られている。 その既知方法を用いて 、 本発明に有用な再構成ヒ ト型抗体を得ることができ、 その好ま し い具体例と して再構成された PM— 1抗体が挙げられる (例えば、 国 際特許出願公開番号 W092- 19759を参照) 。 Furthermore, a reshaped human antibody can be used in the present invention. This is obtained by transplanting the complementarity-determining regions of a mammal other than a human, for example, a mouse antibody, into the complementarity-determining regions of a human antibody. Is also known. Using the known method, a reshaped human antibody useful in the present invention can be obtained, and a preferred example thereof is a reshaped PM-1 antibody (for example, an international patent) (See Application Publication No. W092-19759).
なお、 必要に応じ、 再構成ヒ ト抗体の相補性決定領域が適切な抗 原結合部位を形成するように抗体の可変領域のフ レームワーク (FR ) 領域のア ミ ノ酸を置換してもよい (Satoら、 Cancer Res. 53 : 1 -6, 1993) 。 さ らには抗原に結合し、 IL— 6の活性を阻害するかぎ り、 抗体の断片、 たとえば、 F (ab' )2, Fab あるいは Fv、 H鎖と L鎖の Fvを適当なリ ンカーで連結させたシ ングルチヱイ ン Fv (scFv ) をコー ドする遺伝子を構築し、 これを適当な宿主細胞で発現させ 、 前述の目的に使用することができる。 (例えば、 Birdら、 TIBTEC H, 9 : 132-137, 1991を参照) 。 If necessary, amino acid in the framework (FR) region of the variable region of the antibody may be replaced so that the complementarity determining region of the reshaped human antibody forms an appropriate antigen binding site. Good (Sato et al., Cancer Res. 53: 1-6, 1993). In addition, antibody fragments, such as F (ab ') 2 , Fab or Fv, and Fv of H and L chains, with appropriate linkers, as long as they bind to antigen and inhibit the activity of IL-6 A gene encoding the ligated single-glutinin Fv (scFv) can be constructed, expressed in a suitable host cell, and used for the aforementioned purpose. (See, e.g., Bird et al., TIBTEC H, 9: 132-137, 1991).
scFvは、 抗体の H鎖 V領域と L鎖 V領域を連結してなる。 この sc Fvにおいて、 H鎖 V領域と L鎖 V領域はリ ンカ一、 好ま しく は、 ぺ プチ ドリ ンカ一を介して連結されている (Huston, J. S. ら、 Proc.N atl. Acad. Sci. U. S.A. , 85, 5879-5883, 1988)。 scFvにおける H鎖 V領域および L鎖 V領域は、 上記抗体と して記載されたもののいず れの由来であってもよい。 これらの V領域は、 好ま しく は、 ぺプチ ドリ ンカーによって連結されている。 ぺプチ ドリ ンカーと しては、 例えばァミ ノ酸 12〜19残基からなる任意の一本鎖べプチ ドが用いら scFv is formed by linking the H chain V region and L chain V region of an antibody. In this sc Fv, the H chain V region and the L chain V region are linked via a linker, preferably a peptide linker (Huston, JS et al., Proc. Natl. Acad. Sci. USA, 85, 5879-5883, 1988). The H chain V region and L chain V region in scFv may be derived from any of the antibodies described above. These V regions are preferably connected by peptide linkers. As the peptide linker, for example, any single-chain peptide consisting of amino acid residues 12 to 19 is used.
4レる 4
scFvをコー ドする DNA は、 前記抗体の H鎖または、 H鎖 V領域を コー ドする DNA 、 および L鎖または、 L鎖 V領域をコー ドする DNA を铸型と し、 それらの配列のうちの所望のア ミ ノ酸配列をコー ドす る DNA 部分を、 その両端を規定するプライマ一対を用いて、 PCR 法 により増幅し、 次いで、 さ らにペプチ ドリ ンカ一部分をコー ドする DNA およびその両端を各々 H鎖、 L鎖と連結されるように規定する プライマー対を組み合せて增幅することにより得られる。 The scFv-encoding DNA is a DNA encoding the H chain or the H chain V region of the antibody, and a DNA encoding the L chain or the L chain V region. The DNA portion encoding the desired amino acid sequence is amplified by the PCR method using a pair of primers defining both ends, and then a portion of the peptide linker is also encoded. It is obtained by combining and widening a pair of primers that define DNA and its both ends so as to be linked to an H chain and an L chain, respectively.
また、 一旦 scFvをコー ドする DNA が作成されれば、 それらを含有 する発現ベクター、 および該発現べクターにより形質転換された宿 主を常法に従って得ることができ、 また、 その宿主を用いて常法に 従って、 scFvを得ることができる。 scFvは、 抗体分子に比べ、 組織 への移行性が優れており、 再構成ヒ ト抗体と同様の機能を有するも のと しての利用が期待される。  Further, once DNAs encoding scFv are prepared, an expression vector containing them and a host transformed by the expression vector can be obtained according to a conventional method. The scFv can be obtained according to a conventional method. The scFv is superior to the tissue transferability of the antibody molecule, and is expected to have the same function as the reshaped human antibody.
本発明で使用される IL一 6改変体と しては Brakenhoff ら、 J. Biol . Chem. 269 : 86-93, 1994あるいは Savinoら、 EMBO J. 13 : 1357- 1367, 1994に開示されたものが挙げられる。  The IL-16 variants used in the present invention include those disclosed in Brakenhoff et al., J. Biol. Chem. 269: 86-93, 1994 or Savino et al., EMBO J. 13: 1357-1367, 1994. Is mentioned.
IL— 6改変体と しては、 IL— 6のア ミ ノ酸配列中に置換、 欠失、 揷人といった変異を導入することにより、 IL— 6Rとの結合活性を維 持したまま、 IL一 6のシグナル伝達作用がないものが使用される。 さ らにその由来となる 1L— 6 は上記の性質を有する限り、 その動物 種を問わないが、 抗原性を考慮すればヒ ト由来のものを使用するの が好ま しい。 具体的には、 IL一 6のァ ミ ノ酸配列を公知の分子モデ リ ングプログラム、 たとえば、 WHATIF (Vriendら、 J. Mol. Graphi cs , 8 : 52-56, 1990)を用いてその二次構造を予測し、 さ らに変異ァ ミ ノ酸残基の全体に及ぼす影響を評価することにより行われる。 適当な変異アミ ノ酸残基を決定した後、 ヒ ト IL一 6遗伝子をコー ドする塩基配列を含むベクターを铸型と して、 通常行われる PCR ( ポリ メ レースチェイ ンリアク ショ ン) 法により変異を導入すること により、 1L— 6改変体をコー ドする遺伝子が得られる。 これを必要 に応じて適当な発現ベクターに組み込み、 大腸菌細胞や哺乳類細胞 で発現させ、 培養上清中に含まれたまま、 あるいは通常の手法によ り、 これを単離精製し、 IL一 6Rに対する結合活性および IL— 6のシ グナル伝達の中和活性を評価することができる。 As a modified IL-6, by introducing a mutation such as substitution, deletion, or human mutation into the amino acid sequence of IL-6, the IL-6R can maintain its IL-6R binding activity while maintaining its IL-6R binding activity. One that does not have a signal transduction action is used. Further, 1L-6 as its origin is not limited to animal species as long as it has the above properties, but it is preferable to use human origin in consideration of antigenicity. Specifically, the amino acid sequence of IL-16 can be obtained by using a known molecular modeling program such as WHATIF (Vriend et al., J. Mol. Graphics, 8: 52-56, 1990). This is done by predicting the secondary structure and assessing the overall effect of the mutated amino acid residue. After determining the appropriate mutant amino acid residue, the vector containing the nucleotide sequence encoding the human IL-16 gene is used as a template for PCR (polymerase chain reaction). By introducing a mutation by the method, a gene encoding a 1L-6 variant can be obtained. This is inserted into an appropriate expression vector if necessary, expressed in E. coli cells or mammalian cells, and isolated or purified as it is contained in the culture supernatant or by a conventional method. Binding activity to IL-6 and IL-6 The neutralizing activity of the signal transmission can be evaluated.
本発明で使用される IL一 6部分ペプチ ドあるいは IL— 6R部分ぺプ チ ドは、 各々 IL— 6Rあるいは IL— 6 に結合し、 IL一 6 の活性伝達作 用がないものであれば、 その断片の配列を問わない。 IL— 6部分べ プチ ドおよび IL— 6R部分べプチ ドについては、 米国特許公報 US5210 075 および欧州特許公開公報 ΕΡ6Π126を参照のこと。 IL—6Rアンチ センスオリ ゴヌ ク レオチ ドについては、 特願平 5 — 300338を参照の こ と。  The IL-16 partial peptide or IL-6R partial peptide used in the present invention binds to IL-6R or IL-6, respectively, as long as it has no IL-16 activity transfer action. Regardless of the sequence of the fragment. See US Patent Publication US5210 075 and European Patent Publication No. 6-1126 for IL-6 and IL-6R partial peptides. For the IL-6R antisense oligonucleotide, see Japanese Patent Application No. 5-300338.
本発明の IL— 6 アンタゴニス 卜からなる、 抗腫瘍剤の作用増強剤 は、 1L— 6のシグナル伝達を遮断し、 抗腫瘍剤の作用を捕助、 増強 する限り、 IL一 6Rを有し、 IL— 6 を一つの生理活性物質と して増殖 およびノまたは治療抵抗性を示すいかなる腫瘍の治療に有効に用い るこ とができる。  The antitumor agent potentiator comprising the IL-6 antagonist of the present invention has an IL-16R as long as it blocks 1L-6 signal transduction and captures and enhances the action of the antitumor agent. IL-6 can be effectively used as a physiologically active substance for the treatment of any tumor that shows growth and no resistance or treatment resistance.
本発明の IL一 6 アンタゴニス トからなる、 抗腫瘍剤の作用増強剤 は、 好ま し く は非経口的に、 たとえば、 静脈内注射、 筋肉内注射、 腹腔内注射、 皮下注射等により全身あるいは局部的に投与すること ができる。 さ らに、 少なく と も一種の医薬用担体または希釈剤と と もに医薬組成物ゃキッ 卜の形態をとるこ とができる。  The potentiator of the antitumor agent comprising the IL-16 antagonist of the present invention is preferably administered parenterally, for example, by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, etc. It can be administered steadily. Furthermore, it can be in the form of a pharmaceutical composition kit with at least one pharmaceutical carrier or diluent.
本発明の IL一 6 アンタゴニス トからなる、 抗腫瘍剤の作用増強剤 のヒ 卜に対する投与量は患者の病態、 年齢あるいは投与方法により 異なるが、 適宜適当な量を選択することが必要である。 例えば、 IL -6R抗体の場合、 およそ 1 一 lOOOmgノ患者の範囲で 4 回以下の分割 用量を選択することができる。 また、 1 一 lOmg/kgZ週の用量で投 与するこ とができる。 しかしながら、 本発明の I L— 6 アン夕ゴニス トからなる、 抗腫瘍剤の作用増強剤はこれらの投与量に制限される ものではない。  The dose of the antitumor agent potentiator comprising IL-16 antagonist of the present invention to a human varies depending on the patient's condition, age and administration method, but it is necessary to select an appropriate amount as appropriate. For example, in the case of IL-6R antibody, no more than four divided doses can be selected in a range of approximately 11 mg / l mg of patients. It can also be administered at a dose of 11 mg / kgZweek. However, the dose of the antitumor agent potentiator comprising the IL-6 antagonist of the present invention is not limited to these doses.
本発明の 一 6 アンタゴニス トからなる、 抗腫瘍剤の作用増強剤 は常法にしたがって製剤化することができる (Remington' s Pharma ceut i cal Science, latest edition, Mark Publishing Company, B aston,米国) 。 たとえば、 注射用製剤は、 精製された IL一 6 アン夕 ゴニス トを溶剤、 たとえば、 生理食塩水、 緩衝液、 ブ ドウ糖溶液な どに溶解し、 それに、 吸着防止剤、 たとえば、 Tween80 、 ゼラチン 、 七 ト血清アルブミ ン ( HSA)などを加えたものであり、 または、 使 用前に溶解再構成するために凍結乾燥したものであってもよい。 凍 結乾燥のための賦形剤と しては例えばマ ンニ トール、 ブ ドウ糖など の糖アルコールや糖類を使用することができる。 Antitumor agent action enhancer comprising 16 antagonist of the present invention Can be formulated according to a conventional method (Remington's Pharmaceutical Science, latest edition, Mark Publishing Company, Boston, USA). For example, injectable preparations are prepared by dissolving purified IL-16 gonist in a solvent such as physiological saline, buffer, glucose solution, etc., and adding an anti-adsorption agent such as Tween80, gelatin Or serum albumin (HSA) or lyophilized for reconstitution before use. As an excipient for freeze-drying, for example, sugar alcohols and sugars such as mannitol and glucose can be used.
実施例 Example
以下、 実施例、 参考例および実験例により本発明を具体的に説明 するが、 本発明はこれらに限定されるものではない。  Hereinafter, the present invention will be described specifically with reference to Examples, Reference Examples, and Experimental Examples, but the present invention is not limited thereto.
実施例 1 抗腫瘍剤に対する腫瘍細胞の感受性に関する IL一 6抗 体または IL— 6R抗体の影響  Example 1 Effect of IL-16 Antibody or IL-6R Antibody on Tumor Cell Sensitivity to Antitumor Agent
種々の抗腫瘍剤に対する腫瘍細胞の感受性について、 - 6抗体 または IL— 6R抗体の影響を調べた。  The effect of -6 antibody or IL-6R antibody on the sensitivity of tumor cells to various antitumor agents was examined.
( 1 ) ヒ ト腎細胞癌の調製  (1) Preparation of human renal cell carcinoma
ヒ ト腎細胞癌株 Caki— 1 , Caki - 1 のサブライ ンであるシスブラ チ ン耐性株 Caki— 1 ZDDP 、 ヒ ト肾細胞癌 ACHN、 ヒ ト腎細胞癌 A704 (Giard, D. J. ら、 J. Nat 1. Cancer Inst. 51, 1417-1423, 1973)を 25mM HEPES、 2 mM Lーク"ノレ夕 ミ ン、 1 % non— essent i alア ミ ノ酸 、 100U Zmlペニシ リ ン、 100〃 g /mlス ト レプ ト マイ シ ンおよび 10%加熱不活化ゥ シ胎児血清 (FBS)を含む RPMI 1640 培養液 (以上 、 Gibco 製) (以下、 完全培養液という) 中で、 プラスチッ クディ ッ シュ上で単層になるように培養した。  Human renal cell carcinoma cell lines Caki-1 and Caki-1 are sublines of cis-bratin-resistant cell line Caki-1 ZDDP, human cell carcinoma ACHN, and human renal cell carcinoma A704 (Giard, DJ et al., J. Nat. 1. Cancer Inst. 51, 1417-1423, 1973) was added to 25 mM HEPES, 2 mM L-octylamine, 1% non-essent ial amino acid, 100 U Zml penicillin, 100 g / ml Streptmycin and 10% heat-inactivated. In a RPMI 1640 culture medium (above, manufactured by Gibco) containing fetal serum (FBS) (hereafter referred to as complete culture medium), on a plastic dish. The cells were cultured to form a monolayer.
一方、 Mizutani, Y. ら (Cancer 69, 537-545, 1992) の方法にし たがい、 腎細胞癌患者から新蛘腫瘍細胞を得た。 三人の腎細胞癌患 者の外科的処置の際に、 腎細胞癌腫瘍組織を得た。 組織学的分類に より、 肾細胞癌であることを確認した後、 S mgZmlのコラーゲネー ス (Sigma Chemical Co.製) により、 腫瘍組織を細かく分解し、 腫 瘍細胞懸濁液を調製した。 RPMI 1640 培養液で三回洗浄した後、 細 胞懸濁液を、 15ml容量のプラスチッ クチューブ中の各々 2 mlの 100 %、 80%および 50%の Fico卜 Hypaque からなる非連続的な勾配上に 重層し、 400xg にて 30分間遠心した。 上記 100%の層中のリ ンパ球 に富む単球層を取り除き、 80%の眉から腫瘍細胞と中皮細胞を得た 他細胞の混入を防ぐため、 15mlのブラスチッ クチューブにいれた 完全培養液中で腫瘍細胞に富む細胞懸濁液を、 各々 3 mlの 25%、 15 %、 10%の Percoll からなる非連続的な勾配上に重層して、 室温に て 25xgで 7分間遠心した。 リ ンパ球と分離された腫瘍細胞をチュ一 ブ底面より得た。 得られた腫瘍細胞を洗浄し、 完全培養液で懸濁し た後、 ト リパンブルー染色法により腫瘍細胞の生存を確認した。 こ のように調製したこれらの腫瘍細胞を以下の実験に用いた。 On the other hand, the method of Mizutani, Y. et al. (Cancer 69, 537-545, 1992) was used. Accordingly, neoplastic tumor cells were obtained from renal cell carcinoma patients. Upon surgical treatment of three patients with renal cell carcinoma, renal cell carcinoma tumor tissue was obtained. After confirmation of 肾 cell carcinoma by histological classification, the tumor tissue was finely decomposed with SmgZml collagenase (manufactured by Sigma Chemical Co.) to prepare a tumor cell suspension. After three washes with RPMI 1640 medium, the cell suspension was placed on a discontinuous gradient consisting of 2 ml each of 100%, 80% and 50% fico-hypaque in a 15 ml plastic tube. The layers were overlaid and centrifuged at 400 × g for 30 minutes. Remove the lymphocyte-enriched monocyte layer from the 100% layer and obtain tumor cells and mesothelial cells from 80% eyebrows.To prevent contamination with other cells, complete culture in a 15 ml plastic tube. The cell suspension, enriched in tumor cells, was overlaid on a discontinuous gradient consisting of 3 ml each of 25%, 15%, 10% Percoll and centrifuged at room temperature at 25 × g for 7 minutes. Tumor cells separated from lymphocytes were obtained from the bottom of the tube. After the obtained tumor cells were washed and suspended in a complete culture solution, the survival of the tumor cells was confirmed by trypan blue staining. These tumor cells thus prepared were used in the following experiments.
( 2 ) ELISA 法による腎細胞癌の IL— 6産生確認  (2) Confirmation of IL-6 production in renal cell carcinoma by ELISA
肾細胞癌株 Caki— 1 , Caki - 1 /DDP, ACHN, A704 および肾細胞 癌患者(No. 1 — 3 ) 由来新鮮腫瘍細胞の培養上清中に IL - 6が存在 す.るカヽ否カヽを、 BLISA (enzyme-linked immunosorbent assay; 法で 調べた。  IL IL-6 is present in the culture supernatant of cell tumor cell lines Caki-1 and Caki-1 / DDP, ACHN, A704 and 704 cell carcinoma patients (No. 1 to 3). Was assayed by the BLISA (enzyme-linked immunosorbent assay; method).
96ゥエルの EUSA プレー トに 100〃 1 の IL一 6抗体を添加し、 少 なく ともー晚おいて EL ISA プレー 卜を IL— 6抗体でコー ト した。 こ れらのプレー トは使用するまで 4 °Cにて最大 4週保存した。 IL一 6 抗体でコー ト したプレー トを三回洗浄し、 1 %BSA (ゥシ血清アルブ ミ ン) を含む ELISA PBS で 1 時間ブロ ッ ク した。 二回の洗浄の後、 100 ^ 1 の腫瘍細胞培養上清またはコ ン ト ロールと して大腸菌由来 組換え IL一 6 (Yasukawaら、 Biotechnol. Lett. , 12, 419, 1990) を各ゥヱルに加えた。 A 96-well EUSA plate was supplemented with 100-1 IL-16 antibody, and the ELISA plate was coated with the IL-6 antibody at least. These plates were stored at 4 ° C for up to 4 weeks before use. Plates coated with IL-16 antibody were washed three times and blocked for 1 hour with ELISA PBS containing 1% BSA (perforated serum albumin). After two washes, Escherichia coli-derived recombinant IL-16 (Yasukawa et al., Biotechnol. Lett., 12, 419, 1990) was added to each pellet as a 100 ^ 1 tumor cell culture supernatant or control.
プレー トを 1 時間イ ンキュベー ト し、 三回洗浄して 100〃 1 の抗 一 6 ポリ クローナル抗体 (Matsuda, T. ら、 Eur. J. Immunol., 18 , 951-956, 1988)を各ゥヱルに添加した。 プレー トを 1 時間イ ンキ ュべ一 卜 し、 アルカ リフ ォ スフヱイ ト結合ャギ抗ゥサギ G を各ゥ エルに加え、 さ らに 1 時間ィ ンキュベー ト した。 プレー トを洗浄し 、 ァノレカ リ フ ォ スフ ェ イ ト基質 (Sigma 104, Sigma Chemical Co. 製) とともにィ ンキュペー ト した。 2時間後に EL1SA READER (Immu noreader, Japan Intermed Co. ltd.製) により、 405nm の吸光度を 測定した。 その結果より、 これらの腎細胞癌は IL— 6を産生するこ とが明らかとなった。 (表 1 参照) 。  Plates were incubated for 1 hour, washed three times, and reconstituted with 100 μl of each anti--16 polyclonal antibody (Matsuda, T. et al., Eur. J. Immunol., 18, 951-956, 1988). Was added. Plates were incubated for 1 hour, Alkaline phosphatite-conjugated goat anti-heron G was added to each well and incubated for an additional hour. Plates were washed and incubated with anorecalophosphate substrate (Sigma 104, Sigma Chemical Co.). Two hours later, the absorbance at 405 nm was measured using EL1SA READER (Immunoreader, manufactured by Japan Intermed Co. ltd.). The results revealed that these renal cell carcinomas produced IL-6. (See Table 1).
表 1  table 1
上清中の IL— 6濃度  IL-6 concentration in supernatant
RCC細胞 IL一 6澳度 (pgZml; 平均土標準偏差)RCC cells IL-1-6 degree (pgZml; average soil standard deviation)
Caki - 1 1337±35 Caki-1 1337 ± 35
Caki - 1 DDP 3900± 325  Caki-1 DDP 3900 ± 325
A704 1290土 141  A704 1290 Sat 141
ACHN 1282± 106  ACHN 1282 ± 106
新鮮な RCC 細胞 (患者 No.1) 1236±71  Fresh RCC cells (Patient No. 1) 1236 ± 71
新鮮な RCC 細胞 (患者 No.2) 42±4  Fresh RCC cells (Patient No. 2) 42 ± 4
新鮮な RCC 細胞 (患者 No.3) 2579±219  Fresh RCC cells (Patient No. 3) 2579 ± 219
( 3 ) 抗腫瘍剤の細胞毒性に関する 一 6抗体または IL一 6R抗体の 影響 (3) Effect of 16 antibody or IL-16R antibody on cytotoxicity of antitumor agent
各種濃度の抗腫瘍剤、 すなわち、 シスプラチン (cis-diamminedi chloroplat inum (II)). マイ 卜 マイ シ ン C (mitomycin C; MMC) 、 ア ド リ アマイ シ ン (adriamycin; ADR)、 ビンブラスチ ン (vinblast ine; VBL) および 5 — フノレオロ ウ ラ シノレ ( 5 — f luorouraci 1; 5 ― FU) に対する各腎細胞 Caki— 1 , Caki - 1 /DDP, ACHN, A704 およ び患者(No. 1 - 3 ) 由来新鮮腫瘍細胞の感受性に関する IL - 6抗体 または 1L— 6R抗体の影響を調べるために、 MTT 法 (Mizutani, Y. ら 、 Cancer 73, 730-737, 1994) を実施した。 Various concentrations of antitumor agents, ie, cisplatin (cis-diamminedi chloroplatinum (II)). mitomycin C (MMC), adriamycin (ADR), vinblastine (VBL) and 5 — phnoreolouracinole (5 — IL-6 antibody or 1L on the sensitivity of each renal cell Caki-1, Caki-1 / DDP, ACHN, A704 and fresh tumor cells from the patient (No. 1-3) to F. luorouraci 1; To examine the effect of the 6R antibody, the MTT method (Mizutani, Y. et al., Cancer 73, 730-737, 1994) was performed.
100 1 の上記の腎細胞癌腫瘍細胞懸濁液 ( 2 X 104 細胞) を 96 ウ エノレ底面マイ クロタイタープレー ト (Corning Glass Works, Cor ning製) に分注した。 プレー トを 37°Cにて、 5 %C02 存在加湿下に おき、 腫瘍細胞を 24時間培養した。 細胞培養上清を吸引除去し、 腫 瘍細胞を RPMI 1640 培養液で三回洗浄した。 IL— 6抗体 (Matsuda, T. ら、 Eur. J. Immunol., 18, 951-956, 1988)または I L— 6R抗体 ( Hirata, Y. ら、 J. Immunol., 143, 2900-2906, 1989) との共存下に おいて 200〃 1 の各抗腫瘍剤を含む溶液またはコ ン ト ロールと して 完全培養液を各ゥ ルに添加し、 37°Cにて 24時間培養した。 20^ 1 の MTT 溶液 ( 5 mg/mK Si ma Chemical Co.製) 各ゥエルに加え、 引続き 37°Cで、 5 %C02 存在加湿下にて 4時間培養した。 培養液を 各ゥヱルから取り除き、 0.05N HC1を含むイソプロパノ ール (Sigm a Chemical Co.製) と置換した。 100 1 of the above renal cell carcinoma tumor cell suspension (2 × 10 4 cells) was dispensed into a 96-well bottom microtiter plate (Corning Glass Works, Corning). The plates at 37 ° C, placed under 5% C0 2 present humidified, and the tumor cells were cultured for 24 hours. The cell culture supernatant was aspirated off and the tumor cells were washed three times with RPMI 1640 medium. IL-6 antibody (Matsuda, T. et al., Eur. J. Immunol., 18, 951-956, 1988) or IL-6R antibody (Hirata, Y. et al., J. Immunol., 143, 2900-2906, 1989) ), A complete culture solution was added to each well as a solution or control containing 200% of each antitumor agent, and cultured at 37 ° C for 24 hours. 20 ^ 1 of MTT solution (manufactured by Co. 5 mg / mK Si ma Chemical ) was added to each Ueru, with continued 37 ° C, and incubated for 4 hours at 5% C0 2 present humidified under. The culture solution was removed from each bottle and replaced with isopropanol containing 0.05N HC1 (manufactured by Sigma Chemical Co.).
各ゥェルの溶液の 540nm における吸光度をマイクロカルチユアプ レー ト リーダー ( Immunoreader, Japan Intermed Co. ltd.製) で測 定した。 細胞毒性の割合を次の式にて計算した。 細胞毒性 (%) = The absorbance at 540 nm of each gel solution was measured with a microculture plate reader (Immunoreader, manufactured by Japan Intermed Co. ltd.). The percentage of cytotoxicity was calculated by the following formula. Cytotoxicity (%) =
( 1 - (実験群の吸光度ノコ ン ト ロール群の吸光度) 〕 χιοο 。 その結果、 Caki— 1 細胞では、 抗体との共存下で、 シスブラチ ン (図 1 A, B参照) または MMC (図 2 A, B参照) による細胞毒性が 明らかに上昇した。 コ ン ト ロール抗体 M0PC 3 ZC (J. Natl. Cancer Inst. (Bethesda), 41, 1083, 1968)を加えた実験群では、 シスプ ラチンまたは關 C に対する感受性に影響はみられなかった。 抗腫瘍 剤単独を添加した場合と比べ、 抗腫瘍剤と IL一 6抗体または IL— 6R 抗体の共存下では同程度の細胞毒性の効果を見出すための抗腫瘍剤 の必要量は 1 Z10— 1 Z100 であった。 一方、 IL— 6抗体または IL 一 6R抗体と ADR, VBLまたは 5 — FUの共存下では、 腫瘍細胞の抗腫瘍 剤に対する感受性は変化しなかった。 (1-(Absorbance of experimental group: Absorbance of control group)) χιοο As a result, in Caki-1 cells, cisbratin (see Fig. 1A, B) or MMC (Fig. A, B) clearly increased the cytotoxicity of the control antibody M0PC 3 ZC (J. Natl. Cancer In the experimental group to which Inst. (Bethesda), 41, 1083, 1968) was added, there was no effect on the sensitivity to cissplatin or related C. Compared to the case where the antitumor agent is added alone, the required amount of the antitumor agent is 1 Z10-1 to find the same level of cytotoxic effect in the presence of the antitumor agent and IL-16 antibody or IL-6R antibody. Z100. On the other hand, in the co-presence of IL-6 antibody or IL-16R antibody with ADR, VBL or 5-FU, the sensitivity of tumor cells to antitumor agents did not change.
Caki - 1 /DDP 細胞のシスブラチンに対する耐性は、 IL一 6抗体 または IL—6R抗体とシスブラチンが共存することで克服された (図 3 A, B参照) 。 このような IL— 6抗体または IL— 6R抗体とシスプ ラチンとの共同作用は、 他の肾細胞癌株 ACHN (図 4 A, B参照) 、 A704 (図 5 A, B参照) および三人の患者から得られた新鲜腫瘍細 胞 (図 6, 7, 8の各 A, B参照) においても同様に認められた。  The resistance of Caki-1 / DDP cells to cisplatin was overcome by the coexistence of IL-16 antibody or IL-6R antibody and cisbratin (see FIGS. 3A and B). Such synergy between IL-6 antibody or IL-6R antibody and cisplatin was demonstrated by other 肾 cell carcinoma lines ACHN (see FIGS. 4A and B), A704 (see FIGS. 5A and B) and The same was observed in neoplastic tumor cells obtained from patients (see A and B in Figs. 6, 7, and 8).
実施例 2 カルボブラチンに対する腎細胞癌の感受性に関する IL 一 6抗体または IL一 6R抗体の影響  Example 2 Effect of IL-16 Antibody or IL-16R Antibody on Renal Cell Carcinoma Sensitivity to Carbobratin
腎細胞癌株 Caki— 1 を用い、 各種の濃度に調製したカルボブラチ ン (carboplatin)と IL一 6抗体または 一 6R抗体が共存したときの 細胞毒性について上記実施例 1 と同様に検討した。 その結果、 Caki 一 1細胞のカルボブラチンに対する感受性が増強された (図 9 A, B参照) 。  Using the renal cell carcinoma cell line Caki-1, the cytotoxicity of carboplatin prepared at various concentrations and the IL-16 antibody or the 16R antibody was examined in the same manner as in Example 1 above. As a result, the sensitivity of Caki-11 cells to carbobratin was enhanced (see FIGS. 9A and B).
実施例 3 抗腫瘍剤の細胞内蓄積に関する IL一 6抗体または IL一  Example 3 IL-16 antibody or IL-1 related to intracellular accumulation of antitumor agent
6R抗体の影響  Effect of 6R antibody
10 z g/mlのシスブラチンまたは 100 z gZmlの 5 — FUとコ ン ト ロールと しての培養液、 10 g "mlのコ ン ト ロール抗体 M0PC 3 /C 、 10/z gZmlの 1L一 6抗体または I L一 6R抗体の組合せの存在下で、 Caki - 1 細胞を 24時間培養した。  10 zg / ml cisplatin or 100 z gZml of 5—FU and control culture, 10 g "ml of control antibody M0PC3 / C, 10 / z gZml of 1L-16 antibody Alternatively, Caki-1 cells were cultured for 24 hours in the presence of the IL-16R antibody combination.
次いで、 培養液を取り除き、 細胞を RPMI 1640 培養液で三回洗浄 した。 シスブラチンの細胞内蓄積を、 フ レームレス原子吸光スぺク トロメ 卜 リ ー法 (Daley-Tates, P. T. ら、 Biochem. Pharmacol. , 34 , 2263-2369 ; Riley, C. M. ら、 Analytical Biochem. , 124, 167- 179, 1982)にしたがい測定した。 測定機器は、 ジーマン Z — 8000 ( Zeeman z - 8000 Spectrophotometer, Hitachi Co. ltd.製) を用いた 。 また、 5 — FUの細胞内蓄積を、 ガスク ロマ トグラフ イ ー 質量分 析法 (Marunaka, T., ら、 J. Pharm. Sci., 69, 1296-1300, 1980)で 測定した。 測定機器は、 JGC- 20KPガスク ロマ ト グラフを備えた JMS- D 300 マススぺク ト ロメ ーター (J0EL製) を用いた。 その結果を表 2 に示す。 IL— 6抗体または IL—6R抗体はシスブラチン、 5 — FUの 腫瘍細胞内蓄積に何ら影響を与えないこ とが明らかになった。 The medium is then removed and the cells are washed three times with RPMI 1640 medium did. The intracellular accumulation of cisplatin was determined by the frameless atomic absorption spectrometry method (Daley-Tates, PT et al., Biochem. Pharmacol., 34, 2263-2369; Riley, CM et al., Analytical Biochem., 124, 167). -179, 1982). As a measuring instrument, Zeeman Z-8000 (Zeeman z-8000 Spectrophotometer, manufactured by Hitachi Co. Ltd.) was used. The intracellular accumulation of 5-FU was measured by gas chromatography-mass spectrometry (Marunaka, T., et al., J. Pharm. Sci., 69, 1296-1300, 1980). As a measuring instrument, a JMS-D 300 mass spectrometer (manufactured by J0EL) equipped with a JGC-20KP gas chromatograph was used. The results are shown in Table 2. The IL-6 or IL-6R antibody was found to have no effect on the accumulation of cisplatin, 5-FU in tumor cells.
表 2  Table 2
CDDP又は 5— FUの細胞内蓄積 (ngZlO7 細胞) Intracellular accumulation of CDDP or 5-FU (ngZlO 7 cells)
薬 剤 処 理  Drug treatment
対照 (培養液) 対照 Ab 抗 IL— 6mAb 抗 IL— 6R mAb CDDP 0.28±0.05 0.27±0.02 0.26±0.05 0.27±0.02 5— FU 1.48±0.32 1.57±0.45 1.50±0.19 1.63±0.31 数値は 3回の実験のデータから計算 (平均土標準偏差)  Control (Culture medium) Control Ab Anti-IL-6mAb Anti-IL-6R mAb CDDP 0.28 ± 0.05 0.27 ± 0.02 0.26 ± 0.05 0.27 ± 0.02 5—FU 1.48 ± 0.32 1.57 ± 0.45 1.50 ± 0.19 1.63 ± 0.31 Numerical values of three experiments Calculated from average data (average soil standard deviation)
実施例 4 Glutathione S-transf erase- (GST- π ) 発現に関す  Example 4 Expression of Glutathione S-transf erase- (GST-π)
るシスブラチン、 IL— 6抗体または 一 6R抗体の影響 Caki— 1 細胞を、 コ ン ト ロールと しての培養液、 10 g Zmlの シ スプラチン、 10〃 g /mlの IL— 6抗体または IL一 6R抗体とと もに 4 時間培養した。 ついで、 細胞の全 RNA を Mizutani, Y, ら (Cancer 7 3, 730-737, 1994) の方法により調製し、 10 g RNAZレーンとな るよ う 200mM モップス (M0PS; 3 - 〔 N—モルホ リ ノ〕 プロパンス ルホン酸) 、 50mM酢酸ナ ト リ ゥムおよび 10mM EDTA ナ ト リ ウムを含 む 1 x MOPS緩銜液中で、 1.2%ァガロース— 2.2M HCH0 ゲルで電 気泳動した。 次いで、 RNA を、 3 M NaCl 、 0.3Mクェン酸ナ ト リ ゥム (pH7.0)を含む 20X SSC 溶液中で Biodyne A メ ンブレ ン (Poll 製) に転写した。 50— lOOng の GST— 7Γ cDNA プローブ (Nakagawa , K. ら、 J. Biol. C em. 265, 4296-4301, 1990)を、 な 32 P— dCTP (NEN製) によりラ ンダムオリ ゴブライマー伸長法で標識した。 RNA を転写した上記ナイ ロ ン膜を紫外線でク ロス リ ンク し、 上記プロ一 ブとハイブリ ダィズさせた。 結果を図 10に示す。 Effect of cisplatin, IL-6 antibody or 16R antibody on Caki-1 cells as control medium, 10 g Zml cisplatin, 10 μg / ml IL-6 antibody or IL-1R The cells were cultured with the 6R antibody for 4 hours. Then, the total RNA of the cells was prepared by the method of Mizutani, Y, et al. (Cancer 73, 730-737, 1994), and 200 mM mops (M0PS; 3-[N-morpholy) was used to obtain 10 g RNAZ lanes. [No] propanesulfonic acid), 50 mM sodium acetate and 10 mM EDTA sodium The gel was electrophoresed on a 1.2% agarose-2.2M HCH0 gel in 1x MOPS buffer. Then, the RNA was transferred to Biodyne A membrane (Poll) in a 20X SSC solution containing 3 M NaCl and 0.3 M sodium citrate (pH 7.0). 50—100 ng of GST—7Γ cDNA probe (Nakagawa, K. et al., J. Biol. Cem. 265, 4296-4301, 1990) labeled with 32 P—dCTP (NEN) using the random oligo primer extension method did. The nylon membrane onto which the RNA was transferred was cross-linked with ultraviolet light and hybridized with the probe. The results are shown in FIG.
シスブラチンにより Caki— 1 細胞の GST— 7Γ mRNA の発現は何ら 影響を受けなかった。 しかしながら、 IL- 6抗体または IL— 6R抗体 を添加する と GST— 7Γ mRNA の発現が低下した。 これらのこ とから 、 IL— 6抗体または 一 6R抗体による腎細胞癌のシスブラチンに対 する感受性上昇に関し、 GST— 7Γ mRNA の発現レベルの低下が関与 しているこ とが示唆された。 産業上の利用可能性  Cisplatin did not affect GST-7Γ mRNA expression in Caki-1 cells. However, the addition of IL-6 antibody or IL-6R antibody reduced the expression of GST-7 mRNA. These facts suggest that the decrease in the expression level of GST-7Γ mRNA is involved in increasing the sensitivity of renal cell carcinoma to cisplatin by the IL-6 antibody or the 16R antibody. Industrial applicability
IL- 6抗体または IL— 6R抗体といった IL一 6 アンタゴニス トを抗 腫瘍剤と共存させるこ とで、 より低用量で抗腫瘍剤に対する腫瘍細 胞の感受性が認められ、 IL一 6 アンタゴニス ト と抗腫瘍剤により、 併用結果が発揮されるこ とが確認される。 さ らに、 抗腫瘍剤に治療 抵抗性を示す腫瘍細胞は、 IL一 6 アンタゴニス トにより抗腫瘍剤感 受性を増強される結果、 治療が可能となるこ とが証明される。  By coexisting an IL-16 antagonist such as an IL-6 antibody or an IL-6R antibody with the antitumor agent, the sensitivity of the tumor cell to the antitumor agent was observed at a lower dose, and the IL-16 antagonist was inhibited by the antitumor agent. It is confirmed that the combination results are exhibited by the tumor agent. Furthermore, it is demonstrated that tumor cells that are resistant to treatment with an antitumor agent can be treated as a result of enhancing the sensitivity of the antitumor agent to IL-16 antagonist.
また、 本発明の IL— 6 アンタゴニス トからなる抗腫瘍剤の作用增 強剤は、 抗腫瘍剤の投与必要量を低下させることにより、 抗腫瘍剤 の組織へ及ぼす毒性を低減するこ とができ、 したがって、 抗腫瘍剤 の作用増強剤と して期待される。  In addition, the potentiator of the antitumor agent comprising IL-6 antagonist of the present invention can reduce the toxicity of the antitumor agent to tissues by reducing the required dose of the antitumor agent. Therefore, it is expected as an action enhancer of an antitumor agent.

Claims

請 求 の 範 囲 The scope of the claims
1 . イ ンタ一ロイキン 6 ( IL- 6 ) アンタゴニス トを含んでなる 、 抗腫瘍剤の作用増強剤。 1. An antitumor agent action enhancer comprising interleukin 6 (IL-6) antagonist.
2. IL— 6 アンタゴニス トがヒ ト IL— 6 のアンタゴニス トである 請求項 1 の作用増強剤。  2. The action enhancer according to claim 1, wherein the IL-6 antagonist is human IL-6 antagonist.
3. IL- 6 アンタ ゴニス トがモノ ク ローナル抗体である請求項 1 または 2 の作用増強剤。  3. The action enhancer according to claim 1 or 2, wherein the IL-6 antagonist is a monoclonal antibody.
4. IL— 6 アンタゴニス 卜が 1L— 6抗体または IL一 6 レセプター 抗体である請求項 3 の作用増強剤。  4. The action enhancer according to claim 3, wherein the IL-6 antagonist is a 1L-6 antibody or an IL-16 receptor antibody.
5. 一 6 アンタゴニス トが PM— 1 抗体である請求項 4 の作用増 強剤。  5. The potentiator of claim 4, wherein the one antagonist is a PM-1 antibody.
6. 1L— 6 アンタゴニス トがヒ ト型化 PM— 1 抗体である請求項 5 の作用増強剤。  6. The action enhancer according to claim 5, wherein the 1L-6 antagonist is a humanized PM-1 antibody.
7. 抗腫瘍剤が化学療法剤である請求項 1 ないし 6 の作用増強剤  7. The action enhancer according to any one of claims 1 to 6, wherein the antitumor agent is a chemotherapeutic agent.
8. 抗腫瘍剤が抗腫瘍作用を有する白金化合物である請求項 7 の 作用増強剤。 8. The activity enhancer according to claim 7, wherein the antitumor agent is a platinum compound having an antitumor effect.
9. 抗腫瘍剤がマイ トマィ シン Cである請求項 7 の作用増強剤。 9. The action enhancer according to claim 7, wherein the antitumor agent is mitomycin C.
10. 抗腫瘍剤がシスブラチン、 カルボブラチン、 254- S 、 DWA-21 14R または NK- 121からなる群より選ばれる請求項 8 の作用増強剤。 10. The action enhancer according to claim 8, wherein the antitumor agent is selected from the group consisting of cisbratin, carboblatin, 254-S, DWA-2114R or NK-121.
11. 抗腫瘍作用が増強される腫瘍が、 IL一 6 レセプ夕一を有し、 IL一 6 を生理活性物質と して増殖および Zまたは治療抵抗性を示す 腫瘍である、 請求項 1 ないし 10の作用増強剤。  11. The tumor whose antitumor effect is enhanced is a tumor having IL-16 receptor and showing proliferation and Z or treatment resistance using IL-16 as a bioactive substance. Action enhancer.
PCT/JP1995/002769 1994-12-29 1995-12-28 Antitumor agent potentiator comprising il-6 antagonist WO1996020728A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US08/860,487 US6086874A (en) 1994-12-29 1995-12-28 Antitumor agent effect enhancer containing IL-6 antagonist
ES95942318T ES2170815T5 (en) 1994-12-29 1995-12-28 Use of a PM-1 antibody or an MH166 antibody to enhance the anti-tumor effect of cisplatin or carboplatin
EP95942318A EP0800829B2 (en) 1994-12-29 1995-12-28 Use of a pm-1 antibody or of a mh 166 antibody for enhancing the anti-tumor effect of cisplatin or carboplatin
AT95942318T ATE214602T1 (en) 1994-12-29 1995-12-28 ANTI-TUMOR AGENT AMPLIFIER INCLUDING AN IL-6 ANTAGONIST
KR1019970704446A KR100252743B1 (en) 1994-12-29 1995-12-28 Antitumor agent potentiator comprising il-6 antagonist
CA2209124A CA2209124C (en) 1994-12-29 1995-12-28 Antitumor agent effect enhancer containing il-6 antagonist
DE69525971T DE69525971T3 (en) 1994-12-29 1995-12-28 USE OF A PM-1 ANTIBODY OR AN MH 166 ANTIBODY FOR REINFORCING THE ANTI-TUMOR EFFECT OF CISPLATINE OR CARBOPLATIN
AU43568/96A AU704723B2 (en) 1994-12-29 1995-12-28 Antitumor agent effect enhancer containing IL-6 antagonist
HK98100044A HK1001512A1 (en) 1994-12-29 1998-01-05 Antitumor agent potentiator comprising il-6 antagonist

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP34042594 1994-12-29
JP6/340425 1994-12-29

Publications (1)

Publication Number Publication Date
WO1996020728A1 true WO1996020728A1 (en) 1996-07-11

Family

ID=18336838

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP1995/002769 WO1996020728A1 (en) 1994-12-29 1995-12-28 Antitumor agent potentiator comprising il-6 antagonist

Country Status (12)

Country Link
US (1) US6086874A (en)
EP (1) EP0800829B2 (en)
JP (2) JP4079461B2 (en)
KR (1) KR100252743B1 (en)
CN (1) CN1075387C (en)
AT (1) ATE214602T1 (en)
AU (1) AU704723B2 (en)
CA (1) CA2209124C (en)
DE (1) DE69525971T3 (en)
ES (1) ES2170815T5 (en)
HK (1) HK1001512A1 (en)
WO (1) WO1996020728A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998058674A1 (en) * 1997-06-20 1998-12-30 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Anti-tumour pharmaceutical compositions capable of reducing drug resistance in tumour cells
WO2003037377A1 (en) * 2001-11-02 2003-05-08 Takeda Chemical Industries Ltd Preventives/remedies for proliferative organ diseases, chronic arthritic diseases, hypertrophic scar or keloid
WO2003090779A1 (en) * 2002-04-25 2003-11-06 Chugai Seiyaku Kabushiki Kaisha Remedy for lung cancer
US8529895B2 (en) 2007-10-02 2013-09-10 Chugai Seiyaku Kabushiki Kaisha Method for suppressing the development of graft-versus-host-disease by administering interleukin 6 receptor antibodies
EP2578231B1 (en) 2010-05-28 2022-10-26 Chugai Seiyaku Kabushiki Kaisha Antitumor t cell response enhancer

Families Citing this family (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4503939A (en) * 1983-08-19 1985-03-12 Westinghouse Electric Corp. Elevator system
AU736282B2 (en) 1997-03-21 2001-07-26 Chugai Seiyaku Kabushiki Kaisha A preventive or therapeutic agent for sensitized T cell- mediated diseases comprising IL-6 antagonist as an active ingredient
US20020187150A1 (en) * 1997-08-15 2002-12-12 Chugai Seiyaku Kabushiki Kaisha Preventive and/or therapeutic agent for systemic lupus erythematosus comprising anti-IL-6 receptor antibody as an active ingredient
DE19818044A1 (en) * 1998-04-22 1999-10-28 Klinge Co Chem Pharm Fab Reducing side effects or neutralizing action of carcinostatic or immunosuppressive agents, especially pyridine derivatives, using vitamin PP compounds, e.g. nicotinamide
US6468528B1 (en) * 1999-02-01 2002-10-22 Amgen Canada Inc. Materials and methods to inhibit Hodgkin and Reed Sternberg cell growth
UA80091C2 (en) * 2001-04-02 2007-08-27 Chugai Pharmaceutical Co Ltd Remedies for infant chronic arthritis-relating diseases and still's disease which contain an interleukin-6 (il-6) antagonist
IL161630A0 (en) * 2001-11-06 2004-09-27 Applied Research Systems Methods of treating endometreosis
MXPA04004671A (en) * 2001-11-14 2005-08-25 Johnson & Johnson Anti-il-6 antibodies, compositions, methods and uses.
SI1576112T1 (en) * 2002-01-18 2012-10-30 Zymogenetics Inc Cytokine receptor zcytor17 multimers
DK1475101T3 (en) * 2002-02-14 2011-01-10 Chugai Pharmaceutical Co Ltd Antibody-containing pharmaceutical solutions
EP1572103A4 (en) * 2002-11-15 2008-02-13 Centocor Inc Anti-angiogenic uses of il-6 antagonists
GB2401040A (en) * 2003-04-28 2004-11-03 Chugai Pharmaceutical Co Ltd Method for treating interleukin-6 related diseases
US20050100550A1 (en) * 2003-11-10 2005-05-12 Mohit Trikha Anti-angiogenic uses of IL-6 antagonists
EP3269738A1 (en) 2004-03-24 2018-01-17 Chugai Seiyaku Kabushiki Kaisha Subtypes of humanized antibody against interleukin-6 receptor
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
PE20061324A1 (en) 2005-04-29 2007-01-15 Centocor Inc ANTI-IL-6 ANTIBODIES, COMPOSITIONS, METHODS AND USES
BRPI0611069A2 (en) 2005-05-06 2010-11-09 Zymogenetics Inc monoclonal antibody, humanized antibody, antibody fragment, use of an antagonist, and hybridoma
CN101484575B (en) 2005-06-08 2013-10-02 森托科尔公司 Cellular therapy for ocular degeneration
JP2007091710A (en) * 2005-08-31 2007-04-12 Ishikawajima Harima Heavy Ind Co Ltd Drug, device for inducing drug, magnetic detection device and method for designing drug
WO2008001851A1 (en) 2006-06-28 2008-01-03 Ihi Corporation Drug, drug induction device, magnetic detector and method of designing drug
BRPI0620797A2 (en) * 2005-12-30 2011-11-22 Merck Patent Ges Mit Beschonkter Haftung anti-il-6 antibodies that prevent the binding of il-6 complexed with il-6ralfa to gp130
US8741643B2 (en) 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
US8080248B2 (en) 2006-06-02 2011-12-20 Regeneron Pharmaceuticals, Inc. Method of treating rheumatoid arthritis with an IL-6R antibody
CA2652976C (en) 2006-06-02 2015-08-11 Regeneron Pharmaceuticals, Inc. High affinity antibodies to human il-6 receptor
CN101563365B (en) 2006-08-03 2012-10-31 瓦西尼斯公司 Anti-IL-6 monoclonal antibodies and uses thereof
US20080081041A1 (en) * 2006-09-29 2008-04-03 Jeffrey Nemeth Method of Using IL6 Antagonists with Mitoxantrone for Prostate Cancer
US8404235B2 (en) 2007-05-21 2013-03-26 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
US9701747B2 (en) 2007-05-21 2017-07-11 Alderbio Holdings Llc Method of improving patient survivability and quality of life by anti-IL-6 antibody administration
MX2009012493A (en) * 2007-05-21 2010-01-20 Alder Biopharmaceuticals Inc Novel rabbit antibody humanization methods and humanized rabbit antibodies.
US8178101B2 (en) 2007-05-21 2012-05-15 Alderbio Holdings Inc. Use of anti-IL-6 antibodies having specific binding properties to treat cachexia
US20090238825A1 (en) * 2007-05-21 2009-09-24 Kovacevich Brian R Novel rabbit antibody humanization methods and humanized rabbit antibodies
NZ581596A (en) * 2007-05-21 2012-02-24 Alderbio Holdings Llc Antibodies to il-6 and use thereof
US8252286B2 (en) 2007-05-21 2012-08-28 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US7906117B2 (en) * 2007-05-21 2011-03-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat cachexia, weakness, fatigue, and/or fever
US8062864B2 (en) 2007-05-21 2011-11-22 Alderbio Holdings Llc Nucleic acids encoding antibodies to IL-6, and recombinant production of anti-IL-6 antibodies
US9080145B2 (en) 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
WO2009018453A1 (en) 2007-07-31 2009-02-05 Lifescan, Inc. Differentiation of human embryonic stem cells
CN101246160B (en) * 2007-09-06 2012-11-21 三峡大学 IL-6/sIL-6R combined molecular model established by ELISA method in vitro
CA3123528A1 (en) 2007-11-27 2009-06-04 Lifescan, Inc. Differentiation of human embryonic stem cells to pancreatic cells
PT2796466T (en) 2007-12-07 2018-02-23 Zymogenetics Inc Humanized antibody molecules specific for il-31
PE20091174A1 (en) 2007-12-27 2009-08-03 Chugai Pharmaceutical Co Ltd LIQUID FORMULATION WITH HIGH CONCENTRATION OF ANTIBODY CONTENT
US20090169484A1 (en) 2007-12-28 2009-07-02 Ihi Corporation Iron-salen complex
KR20190057164A (en) 2008-02-21 2019-05-27 얀센 바이오테크 인코포레이티드 Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
US8188235B2 (en) 2008-06-18 2012-05-29 Pfizer Inc. Antibodies to IL-6 and their uses
ES2697798T3 (en) 2008-06-30 2019-01-28 Janssen Biotech Inc Differentiation of pluripotent stem cells
BRPI0919883A2 (en) 2008-10-31 2015-08-11 Centocor Ortho Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine lineage
RU2522001C2 (en) 2008-10-31 2014-07-10 Сентокор Орто Байотек Инк. Differentiation of human embrionic stem cells in line of pancreatic endocrine cells
CA2742786A1 (en) * 2008-11-13 2010-05-20 Femta Pharmaceuticals, Inc. Humanized anti-il-6 antibodies
KR101837080B1 (en) 2008-11-20 2018-03-09 얀센 바이오테크 인코포레이티드 Pluripotent stem cell culture on micro-carriers
WO2010059778A1 (en) 2008-11-20 2010-05-27 Centocor Ortho Biotech Inc. Methods and compositions for cell attachment and cultivation on planar substrates
CN104800195A (en) 2008-11-20 2015-07-29 株式会社Ihi Auto magnetic metal salen complex compound
US9452227B2 (en) * 2008-11-25 2016-09-27 Alderbio Holdings Llc Methods of treating or diagnosing conditions associated with elevated IL-6 using anti-IL-6 antibodies or fragments
US9212223B2 (en) 2008-11-25 2015-12-15 Alderbio Holdings Llc Antagonists of IL-6 to prevent or treat thrombosis
US8337847B2 (en) 2008-11-25 2012-12-25 Alderbio Holdings Llc Methods of treating anemia using anti-IL-6 antibodies
US8992920B2 (en) 2008-11-25 2015-03-31 Alderbio Holdings Llc Anti-IL-6 antibodies for the treatment of arthritis
US8323649B2 (en) * 2008-11-25 2012-12-04 Alderbio Holdings Llc Antibodies to IL-6 and use thereof
US8420089B2 (en) 2008-11-25 2013-04-16 Alderbio Holdings Llc Antagonists of IL-6 to raise albumin and/or lower CRP
JP5317690B2 (en) 2008-12-27 2013-10-16 株式会社東芝 Memory system
WO2011006273A1 (en) * 2009-07-15 2011-01-20 Eva Kovacs-Benke Combination of a human interleukin-6 antagonist and a human interleukin-6 receptor antagonist in therapy of tumour diseases
MX340952B (en) 2009-07-20 2016-07-29 Janssen Biotech Inc Differentiation of human embryonic stem cells.
GB2484873B (en) 2009-07-20 2014-04-30 Janssen Biotech Inc Differentiation of pancreatic endoderm cells.
BR112012001564A2 (en) 2009-07-20 2015-09-01 Janssen Biotech Inc Differentiation of human embryonic stem cells.
KR20160084500A (en) 2009-10-26 2016-07-13 에프. 호프만-라 로슈 아게 Method for the production of a glycosylated immunoglobulin
US9775921B2 (en) 2009-11-24 2017-10-03 Alderbio Holdings Llc Subcutaneously administrable composition containing anti-IL-6 antibody
US9724410B2 (en) 2009-11-24 2017-08-08 Alderbio Holdings Llc Anti-IL-6 antibodies or fragments thereof to treat or inhibit cachexia, associated with chemotherapy toxicity
KR101923537B1 (en) 2009-12-23 2018-11-29 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
RU2610176C2 (en) 2009-12-23 2017-02-08 Янссен Байотек, Инк. Differentiation of human embryonic stem cells
JO3417B1 (en) 2010-01-08 2019-10-20 Regeneron Pharma Stabilized formulations containing anti-interleukin-6 receptor (il-6r) antibodies
MX358451B (en) 2010-03-01 2018-08-20 Janssen Biotech Inc Methods for purifying cells derived from pluripotent stem cells.
CN107338217B (en) 2010-05-12 2021-02-09 詹森生物科技公司 Differentiation of human embryonic stem cells
US9506036B2 (en) 2010-08-31 2016-11-29 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
AU2011296383B2 (en) 2010-08-31 2016-03-10 Janssen Biotech, Inc. Differentiation of pluripotent stem cells
EP2853589B1 (en) 2010-08-31 2017-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
KR102116202B1 (en) 2010-11-08 2020-05-28 제넨테크, 인크. Subcutaneously administered anti-il-6 receptor antibody
CA2818813C (en) 2010-11-23 2020-10-06 Alder Biopharmaceuticals, Inc. Anti-il-6 antibodies for the treatment of oral mucositis
AR087305A1 (en) 2011-07-28 2014-03-12 Regeneron Pharma STABILIZED FORMULATIONS CONTAINING ANTI-PCSK9 ANTIBODIES, PREPARATION METHOD AND KIT
TWI589299B (en) 2011-10-11 2017-07-01 再生元醫藥公司 Compositions for the treatment of rheumatoid arthritis and methods of using same
JO3370B1 (en) 2011-11-10 2019-03-13 Regeneron Pharma Methods of inhibiting tumor growth by antagonizing il-6 receptor
MX2014007744A (en) 2011-12-22 2015-01-12 Janssen Biotech Inc Differentiation of human embryonic stem cells into single hormonal insulin positive cells.
SG11201405052RA (en) 2012-03-07 2014-10-30 Janssen Biotech Inc Defined media for expansion and maintenance of pluripotent stem cells
SG11201408124PA (en) 2012-06-08 2015-01-29 Janssen Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine cells
KR101942769B1 (en) 2012-12-31 2019-01-28 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
BR112015015714A2 (en) 2012-12-31 2017-07-11 Janssen Biotech Inc suspension and agglomeration of human pluripotent cells for differentiation into pancreatic endocrine cells
WO2014105543A1 (en) 2012-12-31 2014-07-03 Janssen Biotech, Inc. Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
RU2674996C2 (en) 2013-07-04 2018-12-14 Ф. Хоффманн-Ля Рош Аг Immunoassay method with interference suppression for determination of antibodies for drugs in serum samples
US9017678B1 (en) 2014-07-15 2015-04-28 Kymab Limited Method of treating rheumatoid arthritis using antibody to IL6R
AU2015259573B2 (en) 2014-05-16 2021-03-18 Janssen Biotech, Inc. Use of small molecules to enhance MAFA expression in pancreatic endocrine cells
CN107922507B (en) 2015-08-18 2022-04-05 瑞泽恩制药公司 anti-PCSK 9 inhibitory antibodies for treating hyperlipidemic patients receiving lipoprotein apheresis
WO2017147169A1 (en) 2016-02-22 2017-08-31 Ohio State Innovation Foundation Chemoprevention using controlled-release formulations of anti-interleukin 6 agents, synthetic vitamin a analogues or metabolites, and estradiol metabolites
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
WO2018170405A1 (en) 2017-03-17 2018-09-20 Ohio State Innovation Foundation Nanoparticles for delivery of chemopreventive agents
US11498969B2 (en) 2019-01-31 2022-11-15 Sanofi Biotechnology Compositions and methods for treating juvenile idiopathic arthritis
WO2020201362A2 (en) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of predicting and preventing cancer in patients having premalignant lesions

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2598666B2 (en) * 1987-10-19 1997-04-09 忠三 岸本 Anti-human BCDF monoclonal antibody
DE3939706C1 (en) * 1989-12-01 1991-03-21 Centre Regional De Transfusion Sanguine, Besancon, Fr
CA2103258A1 (en) * 1991-05-17 1992-11-18 Cory A. Waters Cytokine receptor targeted molecules for treatment of neoplastic cell growth

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
VINK, A JOURNAL OF EXPERIMENTAL MEDICINE, September 1990, Vol. 172, No. 3, pages 997-1000. *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998058674A1 (en) * 1997-06-20 1998-12-30 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti S.P.A. Anti-tumour pharmaceutical compositions capable of reducing drug resistance in tumour cells
WO2003037377A1 (en) * 2001-11-02 2003-05-08 Takeda Chemical Industries Ltd Preventives/remedies for proliferative organ diseases, chronic arthritic diseases, hypertrophic scar or keloid
US7053050B2 (en) 2001-11-02 2006-05-30 Yoshiko Yasuda Preventives/remedies for proliferative organ diseases chronic arthritic diseases, hypertrophic scar or keloid
WO2003090779A1 (en) * 2002-04-25 2003-11-06 Chugai Seiyaku Kabushiki Kaisha Remedy for lung cancer
US8529895B2 (en) 2007-10-02 2013-09-10 Chugai Seiyaku Kabushiki Kaisha Method for suppressing the development of graft-versus-host-disease by administering interleukin 6 receptor antibodies
EP2578231B1 (en) 2010-05-28 2022-10-26 Chugai Seiyaku Kabushiki Kaisha Antitumor t cell response enhancer

Also Published As

Publication number Publication date
EP0800829A4 (en) 1999-12-22
JP4079461B2 (en) 2008-04-23
ES2170815T3 (en) 2002-08-16
CN1174507A (en) 1998-02-25
JP2008063344A (en) 2008-03-21
EP0800829A1 (en) 1997-10-15
ATE214602T1 (en) 2002-04-15
CA2209124C (en) 2010-03-23
AU4356896A (en) 1996-07-24
EP0800829B2 (en) 2012-07-25
DE69525971D1 (en) 2002-04-25
AU704723B2 (en) 1999-04-29
DE69525971T3 (en) 2013-01-10
EP0800829B1 (en) 2002-03-20
HK1001512A1 (en) 1998-06-26
KR100252743B1 (en) 2000-09-01
DE69525971T2 (en) 2002-11-28
JP4812736B2 (en) 2011-11-09
CN1075387C (en) 2001-11-28
US6086874A (en) 2000-07-11
ES2170815T5 (en) 2012-11-14
JPH08231433A (en) 1996-09-10
CA2209124A1 (en) 1996-07-11

Similar Documents

Publication Publication Date Title
WO1996020728A1 (en) Antitumor agent potentiator comprising il-6 antagonist
JP6827415B2 (en) Combination therapy for the treatment of the disease
Mizutani et al. Sensitization of human renal cell carcinoma cells to cis-diamminedichloroplatinum (II) by anti-interleukin 6 monoclonal antibody or anti-interleukin 6 receptor monoclonal antibody
Weisberg et al. Inhibition of wild-type p53-expressing AML by the novel small molecule HDM2 inhibitor CGM097
EP2500360B1 (en) Compositions and methods for diagnosing and treating cancer
TW201838631A (en) Macrocyclic compounds as ros1 kinase inhibitors
US8518888B2 (en) Method of treatment of gastrointestinal-type cancer with antagonistic antibodies to IL-11R
JP2015502958A (en) Combination therapy for the treatment of cancer
KR20150130421A (en) Met-binding agents and uses thereof
EP2506875A1 (en) Methods for treating cancers comprising k-ras mutations
KR102488967B1 (en) Epitope of regulatory T cell surface antigen and an antibody specifically binding to the epitope thereof
CN113194952A (en) Combination of an HDM2-P53 interaction inhibitor and a BCL 2inhibitor and use thereof for treating cancer
CN105051215A (en) Methods of treating pancreatic cancer
JP2003527441A (en) Drugs that block cell cycle and drugs that contain antibodies
CN113396230A (en) Methods of diagnosis and treatment of cancer
Tartarone et al. Beyond conventional: the new horizon of targeted therapy for the treatment of advanced non small cell lung cancer
WO2022036146A1 (en) Diagnostic and therapeutic methods for cancer
Mizejewski Breast Cancer and Cell Cycle Inhibitors (CCIs): Potential Therapeutic Strategies for CCI Cell Tar-geting and Drug Delivery
TWI805542B (en) Peptides and peptidomimetics in combination with t cell activating and/or checkpoint inhibiting agents for cancer treatment
TW202320848A (en) Methods and compositions for treating cancer
US20150023920A1 (en) Novel compositions and methods for preventing or treating cancer metastasis
Scarabel et al. Pharmacogenetics Role of Genetic Variants in Immune-Related Factors: A Systematic Review Focusing on mCRC. Pharmaceutics 2022, 14, 2468
JP2007512303A (en) Combination therapy
WO2023010093A9 (en) Osmr-specific monoclonal antibodies and methods of their use
AU2022286643A1 (en) Compounds for the treatment of glioblastoma

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 95197477.7

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS KE KG KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TT UA UG US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2209124

Country of ref document: CA

Ref document number: 2209124

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 08860487

Country of ref document: US

Ref document number: 1019970704446

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1995942318

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1995942318

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1019970704446

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1019970704446

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 1995942318

Country of ref document: EP